EP3088897A1 - Method for predicting therapeutic effect of biological preparation on rheumatoid arthritis - Google Patents

Method for predicting therapeutic effect of biological preparation on rheumatoid arthritis Download PDF

Info

Publication number
EP3088897A1
EP3088897A1 EP14867880.8A EP14867880A EP3088897A1 EP 3088897 A1 EP3088897 A1 EP 3088897A1 EP 14867880 A EP14867880 A EP 14867880A EP 3088897 A1 EP3088897 A1 EP 3088897A1
Authority
EP
European Patent Office
Prior art keywords
therapy
determining
tocilizumab
value
patient
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
EP14867880.8A
Other languages
German (de)
French (fr)
Other versions
EP3088897B1 (en
EP3088897A4 (en
Inventor
Kazuyuki Yoshizaki
Kazuko Uno
Mitsuhiro IWAHASHI
Katsumi Yagi
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
YOSHIZAKI, KAZUYUKI
Original Assignee
Osaka University NUC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Osaka University NUC filed Critical Osaka University NUC
Publication of EP3088897A1 publication Critical patent/EP3088897A1/en
Publication of EP3088897A4 publication Critical patent/EP3088897A4/en
Application granted granted Critical
Publication of EP3088897B1 publication Critical patent/EP3088897B1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/564Immunoassay; Biospecific binding assay; Materials therefor for pre-existing immune complex or autoimmune disease, i.e. systemic lupus erythematosus, rheumatoid arthritis, multiple sclerosis, rheumatoid factors or complement components C1-C9
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6863Cytokines, i.e. immune system proteins modifying a biological response such as cell growth proliferation or differentiation, e.g. TNF, CNF, GM-CSF, lymphotoxin, MIF or their receptors
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/715Assays involving receptors, cell surface antigens or cell surface determinants for cytokines; for lymphokines; for interferons
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/10Musculoskeletal or connective tissue disorders
    • G01N2800/101Diffuse connective tissue disease, e.g. Sjögren, Wegener's granulomatosis
    • G01N2800/102Arthritis; Rheumatoid arthritis, i.e. inflammation of peripheral joints
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • the present invention relates to a method of predicting and determining a therapeutic effect of a biological formulation on a rheumatoid arthritis patient. More specifically, the present invention relates to a method of predicting and determining a therapeutic effect, such as the level of improvement in a symptom or the possibility of remission, prior to the administration of a biological formulation to a rheumatoid arthritis patient. Furthermore, the present invention relates to a diagnostic agent for predicting and determining a therapeutic effect due to a biological formulation on a rheumatoid arthritis patient.
  • Rheumatoid arthritis is a systemic inflammatory disease, which is predominantly a lesion in the articular synovial membrane. It is estimated that approximately 700,000 people suffer from rheumatoid arthritis in Japan. Many biological formulations that target inflammatory cytokines have been developed for rheumatoid arthritis therapy. In recent years, anti-TNF- ⁇ agents or anti-IL-6 agents, which inhibit TNF- ⁇ or IL-6 action, have been used in clinical practices.
  • Tocilizumab is a humanized IL-6 receptor antibody, which is an agent that causes rheumatoid arthritis to subside by the action of binding to a membrane-binding IL-6 receptor and a soluble IL-6 receptor to suppress IL-6 signaling.
  • etanercept is a fully human soluble TNF/LT ⁇ receptor formulation consisting of a subunit dimer of an extracellular domain of a human tumor necrosis factor II receptor and an Fc region of a human IgG1.
  • Etanercept is an agent that binds to both TNF ⁇ / ⁇ to inhibit signaling to a TNF receptor to cause rheumatoid arthritis to subside.
  • Adalimumab and infliximab are human and chimeric TNF- ⁇ antibodies, which are agents that cause rheumatoid arthritis to subside by specifically binding to excessively produced TNF- ⁇ and inhibiting the binding of TNF- ⁇ to a TNF- ⁇ receptor.
  • Markers for predicting the effectiveness of a biological formulation targeting an inflammatory cytokine on a rheumatoid arthritis patient have been intensively investigated.
  • a method of using a microDNA chip, a method of using a CRP value at baseline as an indicator, a method of using blood soluble ICAMI concentration and CXCL13 concentration as indicators, a method of using leukocyte ADAMT5 gene expression amount as an indicator Non Patent Literature 4
  • a method of comprehensively analyzing genetic polymorphisms Patent Literatures 1 and 2
  • a method of analyzing a genetic mutation of an IL-6 receptor Patent Literature 3
  • Patent Literature 4 a method of analyzing the IL10RB gene, the IRF5 gene, and polymorphisms of the IRF5 gene (Patent Literature 4) has been reported as a method of predicting the therapeutic effectiveness of infliximab on rheumatoid arthritis. Furthermore, a method of comprehensively analyzing genetic polymorphisms (Patent literature 5) has been reported as a method of predicting the therapeutic effectiveness of an anti-TNF- ⁇ agent such as etanercept, adalimumab, or infliximab on rheumatoid arthritis.
  • Patent literature 5 a method of predicting the therapeutic effectiveness of an anti-TNF- ⁇ agent such as etanercept, adalimumab, or infliximab on rheumatoid arthritis.
  • the objective of the present invention is to provide a method of predicting and determining a therapeutic effect (level of improvement in a symptom or possibility of remission) prior to administration of a biological formulation, which is simple and cost-effective, highly versatile and highly accurate. Further objective of the present invention is to provide a diagnostic agent for carrying out the above-described method.
  • the inventors have analyzed the prognostic state of a rheumatoid arthritis patient administered with the biological formulation and the concentrations of cytokines , chemokines and soluble receptors thereof in a serum of the patient prior to administration of the biological formulation in order to solve the problem to discover that a therapeutic effect on a rheumatoid arthritis patient (e.g., level of improvement in a symptom or possibility of remission) can be predicted and determined in a simple and cost-effective manner at any facility with high accuracy, prior to administering a biological formulation targeting an inflammatory cytokine by utilizing the serum concentration of one or more types selected from the group consisting of sgp130, IP-10, sTNFRI, sTNFRII, GM-CSF, IL-1 ⁇ , IL-2, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12, IL-13, IL-15, Eotaxin, VEGF, MCP-1, T
  • Item 1 A method of predicting and determining a therapeutic effect of a biological formulation targeting an inflammatory cytokine on a rheumatoid arthritis patient, characterized in comprising the step of measuring a concentration of at least one type of determination marker selected from the group consisting of sgp130, IP-10, sTNFRI, sTNFRII, GM-CSF, IL-1 ⁇ , IL-2, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12, IL-13, IL-15, Eotaxin, VEGF, MCP-1, TNF- ⁇ , IFN- ⁇ , FGFbasic, PDGF-bb, sIL-6R, and MIP-1 ⁇ in a serum collected from the rheumatoid arthritis patient prior to the administration of the biological formulation.
  • a concentration of at least one type of determination marker selected from the group consisting of sgp130, IP-10, sTNFRI, sTNFRI
  • a therapeutic effect on a rheumatoid arthritis patient can be accurately estimated, and whether rheumatoid arthritis would enter a state of complete remission due to a biological formulation can be determined with high precision, prior to the administration of the biological formulation targeting an inflammatory cytokine.
  • a level of improvement in a symptom for a rheumatoid arthritis patient can be accurately determined prior to the administration of the biological formulation, thus allowing the establishment of a suitable treatment plan, which takes into consideration the therapeutic effect of the biological formulation.
  • the most effective treatment plan can be established for each patient by selecting the optimal biological formulation for each patient.
  • the present invention is beneficial in terms of medical cost containment, sense of security from the prediction of a therapeutic effect and the like.
  • the present invention enables the establishment of a suitable treatment plan based on an accurate prediction of effectiveness of a biological formulation.
  • the present invention does not require complex and time-consuming genetic analysis which lacks versatility.
  • the present invention uses the concentration of a specific cytokine, chemokine, and/or soluble receptor in a serum as an indicator.
  • the present invention can estimate in advance the effectiveness of a biological formulation targeting an inflammatory cytokine for each patient in a simple and cost-effective manner by using an existing method of measurement.
  • the present invention is a method of determining a therapeutic efficacy of a biological formulation targeting an inflammatory cytokine on a rheumatoid arthritis patient, characterized in comprising the step of measuring a concentration of one or more types selected from the group consisting of sgp130, IP-10, sTNFRI, sTNFRII, GM-CSF, IL-1 ⁇ , IL-2, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12, IL-13, IL-15, Eotaxin, VEGF, MCP-1, TNF- ⁇ , IFN- ⁇ , FGFbasic, PDGF-bb, sIL-6R, and MIP-1 ⁇ in a serum collected from the rheumatoid arthritis patient prior to the administration of the biological formulation.
  • a concentration of one or more types selected from the group consisting of sgp130, IP-10, sTNFRI, sTNFRII, GM-
  • the determining method of the present invention is a method of predicting and determining a therapeutic effect of a biological formulation targeting an inflammatory cytokine on a rheumatoid arthritis patient.
  • a biological formulation targeting an inflammatory cytokine is not particularly limited as long as it is a biological formulation used in rheumatoid arthritis therapy. A therapeutic effect can be predicted and determined in accordance with the type of biological formulation to be used in the determining method of the present invention.
  • Examples of a biological formulation targeting an inflammatory cytokine include anti-IL-6 agents, anti-TNF- ⁇ agents and the like.
  • Specific examples of anti-IL-6 agent include humanized anti-IL-6 receptor antibodies, anti-TNF- ⁇ antibodies, human soluble TNF/LT ⁇ receptors consisting of an Fc region of human IgG1 and a subunit dimer of an extracellular domain of a human tumor necrosis factor receptor II, and the like.
  • humanized anti-IL-6 receptor antibodies include tocilizumab. Further, examples of the human soluble TNF/LT ⁇ receptors more specifically include etanercept. Further, examples of the anti-TNF- ⁇ antibodies more specifically include adalimumab and infliximab.
  • Examples of optimal biological formulations thereamong which are applied in the determining method of the present invention include humanized anti-IL-6 receptor antibodies and humanized soluble TNF/LT ⁇ receptors, and still preferably tocilizumab and etanercept.
  • the determining method of the present invention determines whether administration of a biological formulation is effective in a rheumatoid arthritis patient prior to administration of the biological formulation.
  • target rheumatoid arthritis patients in the determining method of the present invention are not particularly limited, as long as it is prior to administration of the biological formulation.
  • DMARDs such as methotrexate
  • past dosing history of anti-cytokine therapy administration of infliximab etanercept, adalimumab, tocilizumab or the like
  • a therapeutic effect due to a biological formulation can be predicted and determined by selecting a desired determination marker in accordance with the past dosing history of the biological formulation in the determining method of the present invention.
  • the determining method of the present invention uses one or two or more types of determination markers selected from the group consisting of sgp130 (soluble gp130), IP-10 (interferon-inducible protein 10), sTNFRI (soluble receptors for tumor necrosis factor type I), sTNFRII (soluble receptors for tumor necrosis factor type II), GM-CSF (granulocyte macrophage colony-stimulating factor), IL-1 ⁇ (interleukin-1 ⁇ ), IL-2 (interleukin-2), IL-5 (interleukin-5), IL-6 (interleukin-6), IL-7 (interleukin-7), IL-8 (interleukin-8), IL-9 (interleukin-9), IL-10 (interleukin-10), IL-12 (interleukin-12), IL-13 (interleukin-13), IL-15 (interleukin-15), Eotaxin, VEGF (vascular endothelial growth factor), MCP-1 (monocyte chemotactic protein-1), TNF-
  • One type of the aforementioned specific cytokine, chemokine, and soluble receptor may be used alone as a determination marker in the determining method of the present invention. However, it is preferable to use two or more types from thereamong in combination as a determination marker, from the viewpoint of predicting and determining a therapeutic effect due to a biological formulation at a higher precision.
  • the determination marker is appropriately selected and used, depending on the therapeutic effect to be predicted and determined, type of biological formulation to be administered, past dosing history of biological formulation or the like. Specific optimal examples of determination marker are shown below for each therapeutic effect to be predicted and determined.
  • level of improvement in symptom after therapy (level of improvement in value of disease activity indicator; value of disease activity indicator prior to therapy - value of disease activity indicator after therapy) is predicted and determined for biological formulation>
  • a naive patient administered with tocilizumab (hereinafter, also referred to as an "tocilizumab therapy naive patient")
  • at least one type selected from the group consisting of IL-7, IL-8, IL-12, IL-13, IP-10, VEGF, IL-1 ⁇ , TNF- ⁇ , and sIL-6R as a determination marker. It is more preferable to use IL-1 ⁇ , IL-7, TNF- ⁇ , and sIL-6R in combination as a determination marker.
  • a switch patient administered with tocilizumab (hereinafter, also referred to as a "tocilizumab therapy switch patient”), it is preferable to use at least one type selected from the group consisting of IL-1 ⁇ , IL-5, IL-6, IL-7, IL-10, IL-12, IL-13, IL-15, FGFbasic, GM-CSF, IFN- ⁇ , TNF- ⁇ , and VEGF as a determination marker.
  • etanercept therapy naive patient For a naive patient administered with etanercept (hereinafter, also referred to as an "etanercept therapy naive patient"), it is preferable to use at least one type selected from the group consisting of IL-6, IP-10, IL-2, IL-13, IL-15, sIL-6R, and sTNFRI as a determination marker. It is more preferable to use a combination of IL-2, IL-15, sIL-6R, and sTNFRI as a determination marker.
  • a tocilizumab therapy naive patient it is preferable to use at least one type selected from the group consisting of sgp130, IL-8, Eotaxin, IP-10, sTNFRI, sTNFRII, IL-6, and VEGF as a determination marker. It is more preferable to use a combination of sgp130, IL-8, Eotaxin, IP-10, sTNFRI, sTNFRII, and IL-6, or a combination of sgp130, IL-8, Eotaxin, IP-10, sTNFRI, sTNFRII, IL-6, and VEGF as a determination marker.
  • a tocilizumab therapy switch patient it is preferable to use at least one type selected from the group consisting of sgp130, IL-1 ⁇ , IL-2, IL-5, IL-15, GM-CSF, IFN- ⁇ , TNF- ⁇ , and IP-10 as a determination marker. It is more preferable to use a combination of sgp130, IP-10, and GM-CSF as a determination marker.
  • IL-9 IL-9
  • IL-6 IL-13
  • TNF- ⁇ TNF- ⁇
  • VEGF vascular endothelial growth factor
  • tocilizumab For patients administered with tocilizumab (including both tocilizumab therapy naive patients and tocilizumab therapy switch patients), it is preferable to use at least one type selected from the group consisting of sgp130, IP-10, sTNFRII, IL-6, IL-7, MCP-1 and IL-1 ⁇ as a determination marker. It is more preferable to use at least sgp130, and even more preferable to use a combination of (i) sgp130, (ii) IP-10, (iii) sTNFRII, and (iv) IL-6, IL-7, MCP-1 or IL-1 ⁇ .
  • a combination of (i) sgp130, (ii) IP-10, (iii) sTNFRII, and (iv) IL-6, IL-7, MCP-1 or IL-1 ⁇ is especially preferable as a determination marker.
  • a combination of (i) sgp130, (ii) IP-10, (iii) sTNFRII, and (iv) IL-6 or IL-1 ⁇ is especially preferable as a determination marker.
  • IL-9 IL-9
  • TNF- ⁇ IL-9
  • VEGF vascular endothelial growth factor
  • PDGF-bb vascular endothelial growth factor
  • MIP-1 ⁇ a determination marker
  • cytokines, chemokines, and soluble receptors used as a determination marker can be measured by a measurement system utilizing an antigen-antibody reaction such as ELISA.
  • ELISA antigen-antibody reaction
  • Such measuring kits are commercially available.
  • the cytokines , chemokines, and soluble receptors can be measured with a known measuring kit by a known method in the determining method of the present invention.
  • a therapeutic effect due to a biological formulation can be predicted and determined based on a measured value of the determination marker.
  • the prediction and determination include a method in which the determination marker is measured in advance for patients in full remission and patients who are not in remission from therapy with a biological formulation; a regression equation of a measured value of the determination marker (explanatory variable) and a therapeutic effect of biological formulation (objective variable) are found by regression analysis; and a measured value of a determination marker of a rheumatoid arthritis patient targeted for determination is applied to said regression equation.
  • a value of serum concentration ( ⁇ g/ml) is preferably used. Further, it is preferable that a regression equation is derived by multiple regression analysis. The objective variable in the above-described regression equation may be appropriately determined based on the therapeutic effect to be predicted and determined.
  • the objective variable when predicting and determining the level of improvement in a symptom after therapy for a biological formulation, the objective variable may be set to "a value obtained by subtracting a value of a disease activity indicator after a predetermined period of therapy from a value of a disease activity indicator prior to therapy" for analysis by multiple linear regression analysis.
  • the objective variable when predicting and determining a value of a disease activity indicator after therapy for a biological formulation, the objective variable may be set to "a value of disease activity indicator after a predetermined period of therapy" for analysis by multiple linear regression analysis.
  • a value of a disease activity indicator examples include a DAS (Disease activity score)-28 value, CDAI (Clinical Disease Activity Index) value, SDAI (Simple Disease Activity Index) value and the like.
  • a DAS-28 value, CDAI value and SDAI value are correlated with one another and reflect a symptom of rheumatoid arthritis.
  • any of such disease activity indicator values may be used in the determining method of the present invention.
  • a disease activity indicator used in the determining method of the present invention is not limited to those exemplified above. Indicators that may be newly advocated in the future can be used.
  • multiple logistic regression analysis may be used for analysis.
  • a value of a disease activity indicator prior to therapy (DAS-28 value, CDAI value, SDAI value or the like) or a result of evaluation by a Boolean method may be utilized as an explanatory variable.
  • determining method of the present invention should not be interpreted to be limited to the following specific methods.
  • a level of improvement in a symptom after therapy due to a biological formulation can be predicted and determined by multiple linear regression analysis while setting an objective variable as "a value obtained by subtracting a value of a disease activity indicator after a predetermined period of therapy from a value of a disease activity indicator prior to therapy" and an explanatory variable as "a measured value of the determination marker".
  • equations (1) and (2) have been discovered as regression equations for predicting and determining a level of improvement in a symptom after 16 weeks of therapy due to a biological formulation (level of improvement in DAS-28 value; DAS-28 value prior to therapy - DAS-28 value after 16 weeks of therapy), separated by the past dosing history of a rheumatoid arthritis patient and type of biological formulation.
  • a level of improvement in a symptom after 16 weeks of therapy can be predicted and determined by finding an objective variable from applying values to one of the following regression equations (1) and (2) depending on the past dosing history of a rheumatoid arthritis patient subjected to determination and type of biological formulation.
  • a level of improvement in a symptom due to a biological formulation is predicted and determined to be large for the patient for larger values of the objective variable calculated by the following regression equation.
  • the regression equations (1) and (2) demonstrate an example of a regression equation used to predict and determine a level of improvement in DAS-28 value after 16 weeks of therapy due to a biological formulation.
  • a level of improvement in a CDAI value or an SDAI value after 16 weeks of therapy due to a biological formulation can naturally be predicted and determined by multiple linear regression analysis by the same method using a CDAI value or SDAI value.
  • regression equations for predicting and determining a level of improvement in a symptom after 16 weeks of therapy are shown in the above-described regression equations (1) and (2).
  • a level of improvement in a symptom before or after 16 weeks of therapy due to the biological formulations can be predicted and determined by multiple linear regression analysis using the same method.
  • equations (3)-(7) have been discovered as regression equations for predicting and determining a DAS-28 value of a symptom after 16 weeks of therapy due to a biological formulation, separated by the past dosing history of a rheumatism patient and type of biological formulation.
  • a DAS-28 value after 16 weeks of therapy can be predicted and determined by finding an objective variable from applying values to one of the following regression equations (3)-(7), depending on the past dosing history of a rheumatoid arthritis patient subjected to determination and type of biological formulation.
  • the objective variable calculated by the following regression equation is 2.3 or less, the patient is predicted and determined to reach remission due to a biological formulation.
  • regression equation (7) does not use a DAS-28 value prior to etanercept administration as an explanatory variable, a DAS-28 value after 16 weeks of therapy can be predicted while eliminating a subjective opinion of a physician. Thus, regression equation (7) is considered preferable over regression equation (6).
  • the regression equations (3)-(7) show examples of a regression equation used to predict and determine a DAS-28 value after 16 weeks of therapy due to a biological formulation.
  • a CDAI value or SDAI value itself after 16 weeks of therapy due to a biological formulation can naturally be predicted and determined by multiple linear regression analysis by the same method using a CDAI value or SDAI value.
  • regression equations for predicting and determining a value of a disease activity indicator after 16 weeks of therapy are shown in the above-described regression equations (3)-(7).
  • a value of disease activity indicator prior to or after 16 weeks of therapy due to the biological formulations can naturally be predicted and determined by multiple linear regression analysis using the same method.
  • equations (8)-(16) have been discovered as regression equations for predicting and determining the possibility of remission (either remission or not in remission) after 16 weeks of therapy due to a biological formulation, separated by the past dosing history of a rheumatoid arthritis patient and type of biological formulation. It is possible to predict and determine whether remission is reached after 16 weeks of therapy by finding the probability (p) of remission after 16 weeks of therapy from applying values to one of the following regression equations (8)-(16) depending on the past dosing history of a rheumatoid arthritis patient subjected to determination and type of biological formulation.
  • the probability of remission estimated from the following regression equations (8) - (16) refers to the probability of a DAS-28 value being 2.3 or less.
  • a p value computed from regression equations (8)-(16) closer to 1 indicates a higher possibility of remission after 16 weeks of therapy.
  • the p value of 0.5 or higher can predict and determine remission and less than 0.5 can predict and determine no remission for convenience's sake.
  • a DAS-28 value of 2.3 is used as the boundary between remission and non-remission to enhance the precision of prediction and determination of remission because a CRP value tends to decrease and DAS-28 value may decreases regardless of inflammation by inhibiting IL-6.
  • the value is set at a lower value of DAS-28 value (2.6), which is generally considered the boundary between remission and non-remission.
  • Determination markers VEGF and PDGF-bb, DAS-28 value prior to etanercept administration is also used as an explanatory variable.
  • p / 1 ⁇ p exp ⁇ 19.058 + 1.390 ⁇ a + ⁇ 2.763 ⁇ E + 4.962 ⁇ Q
  • regression equation (16) does not use a DAS-28 value prior to etanercept administration as an explanatory variable, the possibility of remission can be predicted while eliminating a subjective opinion of a physician. Thus, regression equation (16) is considered preferable over regression equations (14) and (15).
  • the regression equations (6)-(16) show examples of a regression equation for predicting and determining the possibility of remission after 16 weeks of therapy, with a DAS-28 value after 16 weeks of therapy of 2.3 or lower considered remission and the value over 2.3 as non-remission.
  • the possibility of remission after 16 weeks of therapy due to a biological formulation can naturally be predicted and determined by multiple logistic regression analysis with the same method using a CDAI value or SDAI value.
  • regression equations for predicting and determining the possibility of remission after 16 weeks of therapy are shown in the above-described regression equations (6) - (16).
  • the possibility of remission prior to or after 16 weeks of therapy due to a biological formulation can be predicted and determined by multiple logistic regression analysis using the same method.
  • the determining method of the present invention can predict the therapeutic effectiveness of a biological formulation prior to the administration thereof.
  • the method can be utilized in selecting the optimal biological formulation that should be administered prior to starting therapy.
  • cases in which tocilizumab is administered and cases in which etanercept is administered are each predicted by the aforementioned method and a biological formulation with a higher level of improvement is selected, so that an optimal biological formulation can be administered to the patient.
  • a level of improvement in a symptom after therapy with tocilizumab therapy which is predicted by using regression equation (1) is compared to a level of improvement in a symptom after therapy with etanercept therapy, which is predicted by using regression equation (2), so that the biological formulation with a higher level of improvement can be selected as the optimal biological formulation.
  • a DAS-28 value after 16 weeks of therapy of a naive patient cases in which tocilizumab is administered and cases in which etanercept is administered are each predicted by the aforementioned method and a biological formulation with a lower DAS-28 value after 16 weeks of therapy is selected so that an optimal biological formulation can be administered to the patient.
  • a DAS-28 value after 16 weeks of therapy with tocilizumab therapy which is predicted by using one of regression equations (3)-(5) is compared to a DAS-28 value after 16 weeks of therapy with etanercept therapy, which is predicted by using regression equation (6) or (7), so that the biological formulation with a smaller DAS-28 value can be selected as the optimal biological formulation.
  • cases in which tocilizumab is administered and cases in which etanercept is administered are each predicted by the aforementioned method to select a biological formulation with a higher possibility of remission, so that the optimal biological formulation can be administered to the patient.
  • the possibility of remission with tocilizumab therapy which is predicted by using one of regression equations (8) - (11)
  • the possibility of remission which is predicted by using one of regression equations (14)-(16)
  • the biological formulation with a higher possibility of remission can be selected as the optimal biological formulation.
  • the diagnostic agent of the present invention is a diagnostic agent for determining the effectiveness of therapy due to a biological formulation targeting an inflammatory cytokine for a rheumatoid arthritis patient, characterized by comprising a reagent capable of detecting at least one type of determination marker selected from the group consisting of sgp130, IP-10, sTNFRI, sTNFRII, GM-CSF, IL-1 ⁇ , IL-2, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12, IL-13, IL-15, Eotaxin, VEGF, MCP-1, TNF- ⁇ , IFN- ⁇ , FGFbasic, PDGF-bb, sIL-6R, and MIP-1 ⁇ .
  • the determination marker can be measured by a measurement system utilizing an antigen-antibody reaction such as ELISA.
  • an antigen-antibody reaction such as ELISA.
  • reagents capable of detecting the determination marker include antibodies that can specifically bind to the determination marker and fragments thereof. Further, antibodies that can specifically bind to the determination marker may be bound on a suitable support to be provided as an antibody array.
  • the diagnostic agent of the present invention may comprise a reagent (secondary antibody, color producing substance or the like) required for detecting the determination marker by an antigen-antibody reaction.
  • a reagent secondary antibody, color producing substance or the like
  • a rheumatism patient who has not received anti-cytokine therapy administration of infliximab, etanercept, adalimumab, tocilizumab or the like
  • a switch patient a rheumatism patient who has received anti-cytokine therapy in the past.
  • 155 rheumatoid arthritis patients to whom methotrexate therapy was ineffective, were registered at the Higashihiroshima Memorial Hospital from March 2008 to June 2013.
  • 98 patients received therapy with tocilizumab and the remaining 57 patients received therapy with etanercept.
  • 58 patients were naive patients who had not previously received anti-cytokine therapy and 40 patients were switch patients who had previously received anti-cytokine therapy 1-3 times.
  • 49 patients were naive patients who had not previously received anti-cytokine therapy, and the remaining 8 patients were switch patients who had previously received anti-cytokine therapy.
  • Informed consent was obtained prior to receiving a blood sample supply from all patients. Further, the tests were conducted with permission prior to the study from the ethics committee of the Higashihiroshima Memorial Hospital.
  • Table 1 shows the clinical baseline individual group statistics for group of individuals and clinical diagnosis. Further, Figure 1 shows a trial profile of patients receiving therapy with tocilizumab and patients receiving therapy with etanercept. Figures 2-1 to 2-4 show serum concentration of cytokine/chemokine/soluble receptor prior to therapy. 9 na ⁇ ve patients (8 patients who suffered from side effects or other diseases and 1 patient for whom data could not be obtained for the entire 16 weeks) among patients treated with tocilizumab were eliminated. Further, 1 switch patient suffering from a side effect (patient for whom data could not be obtained for the entire 16 weeks) among patients treated with tocilizumab was eliminated.
  • cytokines In order to create a baseline concentration of cytokines, serum was collected from healthy individuals (56 individual; 20 males and 36 females) without a history of suffering from hepatitis C or cancer. The healthy individuals underwent medical examination by the Louis Pasteur Center for Medical Research or the Higashihiroshima Memorial Hospital and informed consent was received in writing from the healthy individuals. The baseline concentration was used to find a distribution pattern of cytokines/chemokines/soluble receptors.
  • cytokines Prior to therapy, concentrations of cytokines, chemokines, and soluble receptors in the serum of rheumatoid arthritis patients were measured.
  • Figure 1 shows clinical results for naive patients and switch patients administered with 8 mg/kg tocilizumab or 50 mg/kg etanercept once every 4 weeks. After 16 weeks of therapy (after 4 administrations), a therapeutic effect was determined based on DAS-28-CRP values and whether the patient is in remission or non-remission. Results for non-remission were further classified into low, medium and high based on DAS-28-CRP values of the patients. DAS-28-ESR values are extensively used to determine the symptom of rheumatoid arthritis patients. However, it is reported that DAS-28-CRP values are almost interchangeable with DAS-28-ESR values and the same results are derived therefrom ( Ann Rheum Dis. 2007, March 407-409 Comparison of Disease Activity Score (DAS)28-erythrocyte Sedimentation rate and DAS-C-reactive protein threshold votes. Inoue E, Yamanaka H , et al.)
  • Figure 3 shows detailed clinical results of each patient shown in Figure 1 , i.e., results of determining DAS-28-CRP values prior to therapy and after 16 weeks of therapy for naive patients who received tocilizumab therapy, switch patients who received tocilizumab therapy, and naive patients who received etanercept therapy.
  • DAS-28-CRP values may be denoted simply as DAS-28 values.
  • cytokine array system Bio-Plex 200, Bio-Rad Laboratories
  • Serum for all patients and healthy individuals were collected by 10 minutes of centrifugation (1600xg). All serum samples were stored at -80° C.
  • Bio-Plex Human Cytokine 27-Plex Panel is configured such that 27 types of cytokines (IL-1 ⁇ , IL-1RA, IL-2, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12 (p70), IL-13, IL-15, IL-17, basic FGF, eotaxin, G-CSF, GM-CSF, IFN- ⁇ , IP-10, MCP-1, MIP-1 ⁇ , MIP-1 ⁇ , PDGF-bb, RANTES, TNF- ⁇ , and VEGF) can be analyzed.
  • cytokines IL-1 ⁇ , IL-1RA, IL-2, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12 (p70), IL-13, IL-15, IL-17, basic FGF, eotaxin, G-CSF, GM-CSF, IFN- ⁇ , IP-10, M
  • sIL-6R sgp130
  • sTNF-RI sTNF-RII
  • sTNF-RII concentrations of cytokines, chemokines, and soluble receptors of 56 healthy individuals were simultaneously measured to find the distribution patterns thereof.
  • Bio-Plex Manager software version 5.0 was used to conduct data collection and analysis.
  • cytokine/chemokine values were analyzed.
  • concentration ( ⁇ g/ml) were directly used for sgp130.
  • Tables 1 and 2 show clinical baseline individual group statistics, clinical diagnosis and cytokine/chemokine/soluble receptor characteristics.
  • Figures 2-1 to 2-4 shows clinical baseline individual group statistics for healthy individuals and rheumatoid arthritis patients with respect to serum concentrations of cytokines/chemokines/soluble receptors prior to therapy.
  • Stage and Class are results of determination with respect to functional classification criteria for rheumatoid arthritis based on Steinbrocker (1949) classification (I-IV; Steinbrocker O et al: Therapeutic criteria in rheumatoid arthritis.
  • Figure 4 shows the relationship between a DAS-28 value prior to therapy and a value obtained from subtracting a DAS-28 value after 16 weeks from the DAS-28 value prior to therapy (PreDAS-28score - 16W DAS-28 score) in a naive patient who has received tocilizumab therapy (PreDAS-28 score). According to Figure 4 , improvement in DAS-28 values was observed after tocilizumab therapy in most naive patients.
  • naive patients and about 23% of switch patients are expected to exhibit remission after tocilizumab therapy.
  • the final symptoms are not identical, some improvement in the symptom is observed in about 45% of naive patients and about 77% of switch patients. Further, about 36.7% of naive patients are expected to exhibit remission after etanercept therapy. In addition, some improvement in the symptom is observed after etanercept therapy in about 60% of the remaining patients.
  • Figures 2-1 to 2-4 show comparisons of baseline individual group statistics of cytokines/chemokines/soluble receptors prior to therapy for healthy individuals and three groups (naive patients who received tocilizumab therapy, switch patients who received tocilizumab therapy, naive patients who received etanercept therapy).
  • serum concentration other than those for sgp130, sIL-6R and sTNFRI in rheumatoid arthritis patients were significantly higher in comparison to healthy individuals.
  • serum concentrations of cytokines/chemokines were lower in naive patients who received etanercept therapy in comparison to naive patients who received tocilizumab therapy.
  • logIL-1 ⁇ , logIL-5, logIL-6, logIL-7, logIL-10, logIL-12, logIL-13, logIL-15, logFGF, logGM-CSF, logIFN- ⁇ , logTNF- ⁇ and logVEGF significantly matched the level of improvement in DAS-28 values.
  • logIL-6 and logIP-10 significantly matched the level of improvement in DAS-28 values for naive patients who receivedetanercept therapy.
  • logIL-1 ⁇ , logIL-2, logIL-5, logIL-15, logGM-CSF, logIFN- ⁇ , logTNF- ⁇ and sgp130 significantly matched DAS-28 values after 16 weeks of therapy.
  • logIL-9 significantly matched DAS-28 values after 16 weeks of therapy for na ⁇ ve patients who received etanercept therapy.
  • regression equation (4) found based on the multiple linear regression analysis shown in Table 7 was used to find a predicted value of DAS-28 value after 16 weeks of therapy in naive patients who received tocilizumab therapy.
  • Figure 5 shows the results of comparing predicted values of DAS-28 values after 16 weeks of therapy calculated by regression equation (4) and actual values of DAS-28 values after 16 weeks of therapy. It was confirmed from the results that DAS-28 values after 16 weeks of therapy estimated from the results of multiple linear regression analysis shown in Table 7 are very consistent with actual values of DAS-28 values after 16 weeks of therapy.
  • regression equation (5) found based on the multiple linear regression analysis shown in Table 8 was used to find a predicted value of DAS-28 value after 16 weeks of therapy in switch patients who received tocilizumab therapy.
  • Figure 6 shows the results of comparing the predicted values of DAS-28 values after 16 weeks of therapy calculated by regression equation (5) and actual values of DAS-28 values after 16 weeks of therapy. It was confirmed from the results that DAS-28 values after 16 weeks of therapy estimated from the results of multiple linear regression analysis shown in Table 8 are very consistent with actual values of DAS-28 values after 16 weeks of therapy.
  • Regression equation (7) found based on the multiple linear regression analysis shown in Table 10 was used to find a predicted value of DAS-28 value after 16 weeks of therapy in naive patients who received etanercept therapy.
  • Figure 7 shows the results of comparing predicted values of DAS-28 values after 16 weeks of therapy calculated by regression equation (7) and actual values of DAS-28 values after 16 weeks of therapy. It was confirmed from the results that DAS-28 values after 16 weeks of therapy can be estimated to a certain extent from the results of multiple linear regression analysis shown in Table 10.
  • naive patients who received etanercept therapy are classified into patients who are predicted to have a higher therapeutic effect when receiving tocilizumab therapy ( Figure 8 a ), patients who are predicted to have barely any difference observed between etanercept therapy and tocilizumab therapy ( Figure 8 b ), and patients who are predicted to have a higher therapeutic effect observed when receiving etanercept therapy ( Figure 8 c ).
  • tocilizumab therapy is estimated to be more effective than etanercept therapy that was actually received.
  • a more effective therapeutic agent can be selected by estimating DAS-28 values due to tocilizumab therapy and etanercept therapy prior to therapy by the present invention.
  • Table 10 shows the results of analyzing data for cytokine/chemokine/soluble receptor concentrations for naive patients and switch patients who received tocilizumab therapy and naive patients who received etanercept therapy. It was found by simple logistic regression analysis that swollen joint count and tender joint count and DAS-28 values were significantly different between complete remission and non-remission groups. Furthermore, sgp130 was significantly different between complete remission and non-remission groups in naive and switch patients who received tocilizumab therapy (Table 11) .
  • Figure 9 shows the results of analyzing the relationship between serum sgp130 concentration and DAS-28 value prior to therapy for remission and non-remission patients. As is clear from Figure 9 , many patients who have reached remission had a high sgp130 concentration.
  • a multivariable model was examined as a prediction biomarker for remission and non-remission by phased multiple forward logistic regression analysis based on serum concentration of cytokines/chemokines/soluble receptors in patients prior to administration of tocilizumab.

Abstract

The objective of the present invention is to provide a method for simply, inexpensively and accurately assessing, before administering a biological preparation, the therapeutic effect thereof (in particular whether there will be a complete response) or the improvement of symptoms in patients having rheumatoid arthritis.
By using at least one serum concentration selected from the group consisting of sgp130, IP-10, sTNFRI, sTNFRII, GM-CSF, IL-1β, IL-2, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12, IL-13, IL-15, Eotaxin, VEGF, MCP-1, TNF-α, IFN-γ, FGF basic, PDGF-bb, sIL-6R and MIP-1α, the therapeutic effect (improvement of symptoms and possibility of response) of an inflammatory cytokine-targeting biological preparation on a patient having rheumatoid arthritis can be predicted in any type of facility in a simple, inexpensive, and highly accurate manner before administering the biological preparation.

Description

    [Technical Field]
  • The present invention relates to a method of predicting and determining a therapeutic effect of a biological formulation on a rheumatoid arthritis patient. More specifically, the present invention relates to a method of predicting and determining a therapeutic effect, such as the level of improvement in a symptom or the possibility of remission, prior to the administration of a biological formulation to a rheumatoid arthritis patient. Furthermore, the present invention relates to a diagnostic agent for predicting and determining a therapeutic effect due to a biological formulation on a rheumatoid arthritis patient.
  • [Background Art]
  • Rheumatoid arthritis is a systemic inflammatory disease, which is predominantly a lesion in the articular synovial membrane. It is estimated that approximately 700,000 people suffer from rheumatoid arthritis in Japan. Many biological formulations that target inflammatory cytokines have been developed for rheumatoid arthritis therapy. In recent years, anti-TNF-α agents or anti-IL-6 agents, which inhibit TNF-α or IL-6 action, have been used in clinical practices.
  • Conventionally, biological formulations targeting an inflammatory cytokine, such as tocilizumab, etanercept, adalimumab, or infliximab, have been used in rheumatoid arthritis therapy. Tocilizumab is a humanized IL-6 receptor antibody, which is an agent that causes rheumatoid arthritis to subside by the action of binding to a membrane-binding IL-6 receptor and a soluble IL-6 receptor to suppress IL-6 signaling. Further, etanercept is a fully human soluble TNF/LTα receptor formulation consisting of a subunit dimer of an extracellular domain of a human tumor necrosis factor II receptor and an Fc region of a human IgG1. Etanercept is an agent that binds to both TNFα/β to inhibit signaling to a TNF receptor to cause rheumatoid arthritis to subside. Adalimumab and infliximab are human and chimeric TNF-α antibodies, which are agents that cause rheumatoid arthritis to subside by specifically binding to excessively produced TNF-α and inhibiting the binding of TNF-α to a TNF-α receptor.
  • For such biological formulations, a certain level of effectiveness in rheumatoid arthritis therapy is verified, while such formulations have disadvantages such as the formulations being expensive and time-intensive for determining a therapeutic effect. In addition, there are certain percentages of cases with no effect, and expression of side effects, such as an infectious disease or an interstitial pneumonia, has been observed in some cases. For this reason, cases where the biological formulation is usable are limited. Thus, if the effectiveness of a biological formulation targeting an inflammatory cytokine can be estimated in advance for each rheumatoid arthritis patient, this would be a boon to rheumatoid arthritis patients and provide contribution to medical business.
  • Markers for predicting the effectiveness of a biological formulation targeting an inflammatory cytokine on a rheumatoid arthritis patient have been intensively investigated. For example, a method of using a microDNA chip, a method of using a CRP value at baseline as an indicator, a method of using blood soluble ICAMI concentration and CXCL13 concentration as indicators, a method of using leukocyte ADAMT5 gene expression amount as an indicator (Non Patent Literature 4), a method of comprehensively analyzing genetic polymorphisms (Patent Literatures 1 and 2), a method of analyzing a genetic mutation of an IL-6 receptor (Patent Literature 3) and the like have been reported as a method of predicting the therapeutic effectiveness of tocilizumab on rheumatoid arthritis. Further, a method of analyzing the IL10RB gene, the IRF5 gene, and polymorphisms of the IRF5 gene (Patent Literature 4) has been reported as a method of predicting the therapeutic effectiveness of infliximab on rheumatoid arthritis. Furthermore, a method of comprehensively analyzing genetic polymorphisms (Patent literature 5) has been reported as a method of predicting the therapeutic effectiveness of an anti-TNF-α agent such as etanercept, adalimumab, or infliximab on rheumatoid arthritis.
  • However, conventional methods of determining a therapeutic effect on rheumatoid arthritis have disadvantages such as: genetic analysis or the like is required, in addition to the operation being complicated; analysis is time and cost-intensive; there is little versatility; proper diagnosis rate is low; and the like. Furthermore, conventional approaches cannot accurately determine whether rheumatoid arthritis can be in full remission prior to the administration of a biological formulation. Thus, conventional approaches have a problem in that an appropriate therapeutic plan which takes into consideration the therapeutic effect thereof cannot be established prior to administration of a biological formulation.
  • Such background conventional techniques elicit a desire for the establishment of a technique for predicting a therapeutic effect of biological formulation administration to a rheumatoid arthritis patient, which is simple and cost-efficient, highly versatile and highly accurate.
  • [Citation List] [Patent Literature]
    • [PTL 1] International Publication No. WO 2011/128096
    • [PTL 2] Japanese Laid-Open Publication No. 2011-182780
    • [PTL 3] International Publication No. WO 2012/41332
    • [PTL 4] Japanese Laid-Open Publication No. 2009-225713
    • [PTL 5] Japanese Laid-Open Publication No. 2010-088432
    [Summary of Invention] [Solution to Problem]
  • The objective of the present invention is to provide a method of predicting and determining a therapeutic effect (level of improvement in a symptom or possibility of remission) prior to administration of a biological formulation, which is simple and cost-effective, highly versatile and highly accurate. Further objective of the present invention is to provide a diagnostic agent for carrying out the above-described method.
  • The inventors have analyzed the prognostic state of a rheumatoid arthritis patient administered with the biological formulation and the concentrations of cytokines , chemokines and soluble receptors thereof in a serum of the patient prior to administration of the biological formulation in order to solve the problem to discover that a therapeutic effect on a rheumatoid arthritis patient (e.g., level of improvement in a symptom or possibility of remission) can be predicted and determined in a simple and cost-effective manner at any facility with high accuracy, prior to administering a biological formulation targeting an inflammatory cytokine by utilizing the serum concentration of one or more types selected from the group consisting of sgp130, IP-10, sTNFRI, sTNFRII, GM-CSF, IL-1β, IL-2, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12, IL-13, IL-15, Eotaxin, VEGF, MCP-1, TNF-α, IFN-γ, FGFbasic, PDGF-bb, sIL-6R, and MIP-1α.
  • More specifically, the inventors have obtained the following knowledge.
    • (1-1) It was discovered through simple linear regression analysis that when therapy is applied by administering tocilizumab to a rheumatism patient who has not received anti-cytokine therapy (administration of infliximab, etanercept, adalimumab, tocilizumab or the like) in the past (may also be referred to as a "naïve patient" hereinafter), the level of improvement in the DAS-28 value (DAS-28 value prior to therapy - DAS-28 value after 16 weeks of therapy) is significantly correlated with the log values of serum concentrations of IL-7, IL-8, IL-12, IL-13, IP-10, and VEGF prior to the administration of tocilizumab to the patient.
    • (1-2) It was discovered through simple linear regression analysis that when therapy is applied by administering tocilizumab to a rheumatism patient who has received anti-cytokine therapy in the past (also referred to as a "switch patient" hereinafter), the level of improvement in the DAS-28 value is significantly correlated with the log values of serum concentrations of IL-1β, IL-5, IL-6, IL-7, IL-10, IL-12, IL-13, IL-15, FGFbasic, GM-CSF, IFN-γ, TNF-α, and VEGF prior to the administration of tocilizumab to the patient.
    • (1-3) It was discovered through simple linear regression analysis that when therapy is applied by administering etanercept to a naive patient, the level of improvement in DAS-28 value is significantly correlated with the log values of serum concentrations of IL-6 and IP-10 prior to the administration of etanercept to the patient.
    • (1-4) It was discovered through multiple linear regression analysis that when therapy is applied by administering tocilizumab to a naive patient, the level of improvement in DAS-28 value is significantly correlated with a combination of log values of serum concentrations of IL-1β, IL-7, TNF-α, and sIL-6R prior to the administration of tocilizumab to the patient.
    • (1-5) It was discovered through multiple linear regression analysis that when therapy is applied by administering etanercept to a naive patient, the level of improvement in DAS-28 value is significantly correlated with a combination of log values of serum concentrations of IL-2, IL-15, sIL-6R, and sTNFRI prior to the administration of etanercept to the patient.
    • (2-1) It was discovered through simple linear regression analysis that when therapy is applied by administering tocilizumab to a naive patient, the DAS-28 value after 16 weeks of therapy is significantly correlated with a serum concentration of sgp130 prior to the administration of tocilizumab to the patient.
    • (2-2) It was discovered through simple linear regression analysis that when therapy is applied by administering tocilizumab to a switch patient, the DAS-28 value after 16 weeks of therapy is significantly correlated with the log values of serum concentrations of It-1β, IL-2, IL-5, IL-15, GM-CSF, IFN-γ, and TNF-α and a serum concentration of sgp130 prior to the administration of tocilizumab to the patient.
    • (2-3) It was discovered through simple linear regression analysis that when therapy is applied by administering etanercept to a naive patient, the DAS-28 value after 16 weeks of therapy is significantly correlated with the log value of a serum concentration of IL-9 prior to the administration of etanercept to the patient.
    • (2-4) It was discovered through multiple linear regression analysis that when therapy is applied by administering tocilizumab to a naive patient, the DAS-28 value after 16 weeks of therapy is significantly correlated with a combination of log values of serum concentrations of IL-8, Eotaxin, IP-10, sTNRFI, sTNFRII, IL-6 and VEGF and a serum concentration of sgp130 prior to the administration of tocilizumab to the patient.
    • (2-5) It was discovered through multiple linear regression analysis that when therapy is applied by administering tocilizumab to a naive patient, the DAS-28 value after 16 weeks of therapy is significantly correlated with a combination of log values of serum concentrations of IL-8, Eotaxin, IP-10, sTNFRI, sTNRFII, and IL-6 and a serum concentration of sgp130 prior to the administration of tocilizumab to the patient.
    • (2-6) It was discovered through multiple linear regression analysis that when therapy is applied by administering tocilizumab to a switch patient, the DAS-28 value after 16 weeks of therapy is significantly correlated with a combination of log values of serum concentrations of IP-10 and GM-CSF and a serum concentration of sgp130 prior to the administration of tocilizumab to the patient.
    • (2-7) It was discovered through multiple linear regression analysis that when therapy is applied by administering etanercept to a naive patient, the DAS-28 value after 16 weeks of therapy is significantly correlated with a combination of log values of serum concentrations of IL-6 and IL-13 and the DAS-28 value prior to the administration of etanercept.
    • (2-8) It was discovered through multiple linear regression analysis that when therapy is applied by administering etanercept to a naive patient, the DAS-28 value after 16 weeks of therapy is significantly correlated with a combination of log values of serum concentrations of IL-9, TNF-α, and VEGF prior to the administration of etanercept.
    • (3-1) It was discovered through multiple logistic regression analysis that the possibility of remission, when therapy is applied by administering tocilizumab to a naïve patient can be predicted and determined by combining a serum concentration of sgp130, a log value of a serum concentration of IP-10, a log value of a serum concentration of sTNFRII, and a log value of a serum concentration of IL-6, IL-7, MCP-1, or IL-1β prior to the administration of tocilizumab.
    • (3-2) It was discovered through multiple logistic regression analysis that the possibility of remission, when therapy is applied by administering tocilizumab to a switch patient, can be predicted and determined by combining a serum concentration of sgp130, a log value of a serum concentration of IP-10, a log value of a serum concentration of sTNFRII, and a log value of a serum concentration of IL-6 or IL-1β prior to the administration of tocilizumab.
    • (3-3) It was discovered through multiple logistic regression analysis that the possibility of remission, when therapy is applied by administering etanercept to a naive patient, can be predicted and determined by combining the DAS-28 value and log values of serum concentrations of VEGF and PDGF-bb prior to the administration of etanercept.
    • (3-4) It was discovered through multiple logistic regression analysis that the possibility of remission, when therapy is applied by administering etanercept to a naive patient, can be predicted and determined by combining the DAS-28 value and log values of serum concentrations of MIP-1α and PDGF-bb prior to the administration of etanercept.
    • (3-5) It was discovered through multiple linear regression analysis that the possibility of remission, when therapy is applied by administering etanercept to a naive patient, can be predicted and determined by combining log values of serum concentrations of IL-9 and TNF-α prior to the administration of etanercept.
  • The present invention was completed by additional repeated examinations based on such knowledge. Specifically, the present invention provides inventions in the following embodiments. Item 1. A method of predicting and determining a therapeutic effect of a biological formulation targeting an inflammatory cytokine on a rheumatoid arthritis patient, characterized in comprising the step of measuring a concentration of at least one type of determination marker selected from the group consisting of sgp130, IP-10, sTNFRI, sTNFRII, GM-CSF, IL-1β, IL-2, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12, IL-13, IL-15, Eotaxin, VEGF, MCP-1, TNF-α, IFN-γ, FGFbasic, PDGF-bb, sIL-6R, and MIP-1α in a serum collected from the rheumatoid arthritis patient prior to the administration of the biological formulation.
    • Item 2. The method of item 1 of predicting and determining a possibility of remission with tocilizumab, wherein
      the determination marker is at least one type selected from the group consisting of sgp130, IP-10, sTNFRII, IL-6, IL-7, MCP-1, and IL-1β.
    • Item 3. The method of determining of item 2, wherein at least sgp130 is used as the determination marker.
    • Item 4. The method of determining of item 2 or 3, wherein
      a patient to be administered with tocilizumab is a rheumatoid arthritis patient who has not received anti-cytokine therapy in the past, and
      the determination marker is a combination of (i) sgp130, (ii) IP-10, (iii) sTNFRII, and (iv) IL-6, IL-7, MCP-1 or IL-1β. Item 5. The method of determining of item 2 or 3, wherein
      a patient to be administered with tocilizumab is a rheumatoid arthritis patient who has received anti-cytokine therapy in the past, and
      the determination marker is a combination of (i) sgp130, (ii) IP-10, (iii) sTNFRII, and (iv) IL-6 or IL-1β.
    • Item 6. The method of determining of item 1, wherein
      the method is a method of predicting and determining a possibility of remission with etanercept in a rheumatism patient who has not received anti-cytokine therapy in the past, and
      the determination marker is at least one type selected from the group consisting of IL-9, TNF-α, VEGF, PDGF-bb, and MIP-1α. Item 7. The method of determining of item 6, wherein the determination marker is a combination of IL-9 and TNF-α, a combination of VEGF and PDGF-bb, or a combination of MIP-1α and PDGF-bb.
    • Item 8. The method of determining of item 1, wherein
      the method is a method of predicting and determining a disease activity indicator after therapy with tocilizumab in a rheumatism patient who has not received anti-cytokine therapy in the past, and
      the determination marker is at least one type selected from the group consisting of sgp130, IL-8, Eotaxin, IP-10, sTNFRI, sTNFRII, IL-6, and VEGF.
    • Item 9. The method of determining of item 8, wherein the determination marker is a combination of sgp130, IL-8, Eotaxin, IP-10, sTNFRI, sTNFRII, and IL-6 or a combination of sgp130, IL-8, Eotaxin, IP-10, sTNFRI, sTNFRII, IL-6 and VEGF.
    • Item 10. The method of determining of item 1, wherein
      the method is a method of predicting and determining a value of a disease activity indicator after therapy with tocilizumab in a rheumatism patient who has received anti-cytokine therapy in the past, and
      the determination marker is at least one type selected from the group consisting of sgp130, IL-1β, IL-2, IL-5, IL-15, GM-CSF, IFN-γ, TNF-α, and IP-10.
    • Item 11. The method of determining of item 10, wherein the determination marker is a combination of sgp130, IP-10, and GM-CSF.
    • Item 12. The method of determining of item 1, wherein
      the method is a method of predicting and determining a value of a disease activity indicator after therapy with etanercept in a rheumatism patient who has not received anti-cytokine therapy in the past, and
      the determination marker is at least one type selected from the group consisting of IL-9, IL-6, IL-13, TNF-α, and VEGF. Item 13. The method of determining of item 12, wherein
      the determination marker is a combination of IL-9, TNF-α and VEGF or a combination of IL-6 and IL-13.
    • Item 14. The method of determining of item 1, wherein
      the method is a method of predicting and determining a level of improvement in a symptom after therapy with tocilizumab in a rheumatism patient who has not received anti-cytokine therapy in the past, and
      the determination marker is at least one type selected from the group consisting of IL-7, IL-8, IL-12, IL-13, IP-10, VEGF, IL-1β, TNF-α, and sIL-6R.
    • Item 15. The method of determining of item 14, wherein the determination marker is a combination of IL-1β, IL-7, TNF-α, and sIL-6R.
    • Item 16. The method of determining of item 1, wherein
      the method is a method of predicting and determining a level of improvement in a symptom after therapy with tocilizumab in a rheumatism patient who has received anti-cytokine therapy in the past, and
      the determination marker is at least one type selected from the group consisting of IL-1β, IL-5, IL-6, IL-7, IL-10, IL-12, IL-13, IL-15, FGFbasic, GM-CSF, IFN-γ, TNF-α, and VEGF. Item 17. The method of determining of item 1, wherein
      the method is a method of predicting and determining a level of improvement in a symptom after therapy with etanercept in a rheumatism patient who has not received anti-cytokine therapy in the past, and
      the determination marker is at least one type selected from the group consisting of IL-6, IP-10, IL-2, IL-13, IL-15, sIL-6R, and sTNFRI.
    • Item 18. The method of determining of item 17, wherein the determination marker is a combination of IL-2, IL15, sIL-6R, and sTNFRI or a combination of IL-6 and IL-13.
    • Item 19. A method of selecting a more effective biological formulation for therapy in a rheumatism patient who has not received anti-cytokine therapy in the past from among biological formulations consisting of tocilizumab and etanercept, comprising:
      • predicting and determining a possibility of remission with tocilizumab in accordance with the method of determining of item 4;
      • predicting and determining a possibility of remission with etanercept in accordance with the method of determining of item 6; and
      • comparing the possibility of remission with tocilizumab with the possibility of remission with etanercept that were predicted and determined in the aforementioned steps to select a biological formulation with a high possibility of remission. Item 20. A method of selecting a more effective biological formulation for therapy in a rheumatism patient who has not received anti-cytokine therapy in the past from among biological formulations consisting of tocilizumab and etanercept, comprising:
        • predicting and determining a disease activity indicator after therapy with tocilizumab in accordance with the method of determining of item 10 or 11;
        • predicting and determining a disease activity indicator after therapy with etanercept in accordance with the method of determining of item 12 or 13; and
        • comparing the disease activity indicator after therapy with tocilizumab with the disease activity indicator after therapy with etanercept that were predicted and determined in the aforementioned steps to select a biological formulation with a low disease activity indicator after therapy.
    • Item 21. A method of selecting a more effective biological formulation for therapy in a rheumatism patient who has not received anti-cytokine therapy in the past from among biological formulations consisting of tocilizumab and etanercept, comprising:
      • predicting and determining a level of improvement in a symptom after therapy with tocilizumab in accordance with the method of determining of item 14 or 15;
      • predicting and determining a level of improvement in a symptom after therapy with etanercept in accordance with the method of determining of item 17 or 18; and
      • comparing the level of improvement in a symptom after therapy with tocilizumab with the level of improvement in a symptom after therapy with etanercept that were predicted in the aforementioned steps to select a biological formulation with a high level of improvement in a symptom after therapy. Item 22. A diagnostic agent for predicting and determining a therapeutic effect due to a biological formulation targeting an inflammatory cytokine on a rheumatoid arthritis patient, comprising a reagent capable of detecting at least one type of marker selected from the group consisting of sgp130, IP-10, sTNFRI, sTNFRII, GM-CSF, IL-1β, IL-2, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12, IL-13, IL-15, Eotaxin, VEGF, MCP-1, TNF-α, IFN-γ, FGFbasic, PDGF-bb, sIL-6R, and MIP-1α.
    [Advantageous Effects of Invention]
  • Accordingly to the present invention, a therapeutic effect on a rheumatoid arthritis patient can be accurately estimated, and whether rheumatoid arthritis would enter a state of complete remission due to a biological formulation can be determined with high precision, prior to the administration of the biological formulation targeting an inflammatory cytokine. Furthermore, according to the present invention, a level of improvement in a symptom for a rheumatoid arthritis patient can be accurately determined prior to the administration of the biological formulation, thus allowing the establishment of a suitable treatment plan, which takes into consideration the therapeutic effect of the biological formulation. Further, according to the present invention, it is possible to predict which biological formulation is the most effective when administered for a rheumatoid arthritis patient prior to therapy. Thus, the most effective treatment plan can be established for each patient by selecting the optimal biological formulation for each patient.
  • In this manner, a rheumatoid arthritis patient for whom administration of a biological formulation is effective can be identified by utilizing the present invention. Thus, for patients, the present invention is beneficial in terms of medical cost containment, sense of security from the prediction of a therapeutic effect and the like. For physicians, the present invention enables the establishment of a suitable treatment plan based on an accurate prediction of effectiveness of a biological formulation.
  • Furthermore, the present invention does not require complex and time-consuming genetic analysis which lacks versatility. In addition, the present invention uses the concentration of a specific cytokine, chemokine, and/or soluble receptor in a serum as an indicator. Thus, the present invention can estimate in advance the effectiveness of a biological formulation targeting an inflammatory cytokine for each patient in a simple and cost-effective manner by using an existing method of measurement.
  • [Brief Description of Drawings]
    • [Figure 1] Figure 1 is a diagram showing trial profiles of tocilizumab therapy patients and etanercept therapy patients.
    • [Figure 2] Figures 2-1 to 2-4 are diagrams showing clinical baseline individual group statistics for healthy individuals and rheumatoid arthritis patients with respect to serum concentrations of cytokines/chemokines/soluble receptors.
    • [Figure 3] Figure 3 is a diagram showing the relationship between DAS-28 values prior to therapy and DAS-28 values after 16 weeks of therapy in tocilizumab therapy patients and etanercept therapy patients.
    • [Figure 4] Figure 4 is a diagram showing the relationship between DAS-28 values prior to therapy (PreDAS-28 score) and values obtained from subtracting a DAS-28 value after 16 weeks from a DAS-28 value prior to therapy (PreDAS-28score - 16W DAS-28 score) in naive patients who received tocilizumab therapy.
    • [Figure 5] Figure 5 is a diagram showing results of comparing predicted DAS-28 values after 16 weeks of therapy calculated from regression equation (4) prior to therapy and actual DAS-28 values after 16 weeks of therapy subjecting naive patients who received tocilizumab therapy.
    • [Figure 6] Figure 6 is a diagram showing results of comparing predicted DAS-28 values after 16 weeks of therapy calculated from regression equation (5) prior to therapy and actual DAS-28 values after 16 weeks of therapy subjecting switch patients who received tocilizumab therapy.
    • [Figure 7] Figure 7 is a diagram showing results of comparing predicted DAS-28 values after 16 weeks of therapy calculated from regression equation (7) prior to therapy and actual DAS-28 values after 16 weeks of therapy subjecting naive patients who received etanercept therapy.
    • [Figure 8] Figure 8 is a diagram showing the relationship between actual values of DAS-28 after 16 weeks of etanercept therapy and predicted DAS-28 values after 16 weeks of therapy estimated by assuming a patient has received tocilizumab therapy in naive patients who received etanercept therapy.
    • [Figure 9] Figure 9 is a diagram showing results of analyzing the relationship between serum sgp130 concentrations and DAS-28 values prior to therapy for patients in remission and non-remission.
    [Description of Embodiments] 1. Determining method
  • The present invention is a method of determining a therapeutic efficacy of a biological formulation targeting an inflammatory cytokine on a rheumatoid arthritis patient, characterized in comprising the step of measuring a concentration of one or more types selected from the group consisting of sgp130, IP-10, sTNFRI, sTNFRII, GM-CSF, IL-1β, IL-2, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12, IL-13, IL-15, Eotaxin, VEGF, MCP-1, TNF-α, IFN-γ, FGFbasic, PDGF-bb, sIL-6R, and MIP-1α in a serum collected from the rheumatoid arthritis patient prior to the administration of the biological formulation. Hereinafter, the determining method of the present invention is discussed in detail.
  • Biological formulation subjected to determination
  • The determining method of the present invention is a method of predicting and determining a therapeutic effect of a biological formulation targeting an inflammatory cytokine on a rheumatoid arthritis patient.
  • A biological formulation targeting an inflammatory cytokine is not particularly limited as long as it is a biological formulation used in rheumatoid arthritis therapy. A therapeutic effect can be predicted and determined in accordance with the type of biological formulation to be used in the determining method of the present invention. Examples of a biological formulation targeting an inflammatory cytokine include anti-IL-6 agents, anti-TNF-α agents and the like. Specific examples of anti-IL-6 agent include humanized anti-IL-6 receptor antibodies, anti-TNF-α antibodies, human soluble TNF/LTα receptors consisting of an Fc region of human IgG1 and a subunit dimer of an extracellular domain of a human tumor necrosis factor receptor II, and the like. More specific examples of the humanized anti-IL-6 receptor antibodies include tocilizumab. Further, examples of the human soluble TNF/LTα receptors more specifically include etanercept. Further, examples of the anti-TNF-α antibodies more specifically include adalimumab and infliximab.
  • Examples of optimal biological formulations thereamong which are applied in the determining method of the present invention include humanized anti-IL-6 receptor antibodies and humanized soluble TNF/LTα receptors, and still preferably tocilizumab and etanercept.
  • Patients subjected to determination
  • The determining method of the present invention determines whether administration of a biological formulation is effective in a rheumatoid arthritis patient prior to administration of the biological formulation.
  • Further, target rheumatoid arthritis patients in the determining method of the present invention are not particularly limited, as long as it is prior to administration of the biological formulation. In addition, whether DMARDs such as methotrexate are administered, past dosing history of anti-cytokine therapy (administration of infliximab etanercept, adalimumab, tocilizumab or the like) are not relevant. A therapeutic effect due to a biological formulation can be predicted and determined by selecting a desired determination marker in accordance with the past dosing history of the biological formulation in the determining method of the present invention.
  • Determination markers
  • The determining method of the present invention uses one or two or more types of determination markers selected from the group consisting of sgp130 (soluble gp130), IP-10 (interferon-inducible protein 10), sTNFRI (soluble receptors for tumor necrosis factor type I), sTNFRII (soluble receptors for tumor necrosis factor type II), GM-CSF (granulocyte macrophage colony-stimulating factor), IL-1β (interleukin-1β), IL-2 (interleukin-2), IL-5 (interleukin-5), IL-6 (interleukin-6), IL-7 (interleukin-7), IL-8 (interleukin-8), IL-9 (interleukin-9), IL-10 (interleukin-10), IL-12 (interleukin-12), IL-13 (interleukin-13), IL-15 (interleukin-15), Eotaxin, VEGF (vascular endothelial growth factor), MCP-1 (monocyte chemotactic protein-1), TNF-α (tumor necrosis factor-α), IFN-γ (interferon-γ), FGFbasic (basic fibroblast growth factor), PDGF-bb (platelet-derived growth factor bb), sIL-6R (soluble receptors for interleukin-6), and MIP-1α (macrophage inflammatory protein-1α) in the serum of the rheumatoid arthritis patient.
  • One type of the aforementioned specific cytokine, chemokine, and soluble receptor may be used alone as a determination marker in the determining method of the present invention. However, it is preferable to use two or more types from thereamong in combination as a determination marker, from the viewpoint of predicting and determining a therapeutic effect due to a biological formulation at a higher precision.
  • The determination marker is appropriately selected and used, depending on the therapeutic effect to be predicted and determined, type of biological formulation to be administered, past dosing history of biological formulation or the like. Specific optimal examples of determination marker are shown below for each therapeutic effect to be predicted and determined.
  • <Cases where level of improvement in symptom after therapy (level of improvement in value of disease activity indicator; value of disease activity indicator prior to therapy - value of disease activity indicator after therapy) is predicted and determined for biological formulation>
  • For a naive patient administered with tocilizumab (hereinafter, also referred to as an "tocilizumab therapy naive patient"), it is preferable to use at least one type selected from the group consisting of IL-7, IL-8, IL-12, IL-13, IP-10, VEGF, IL-1β, TNF-α, and sIL-6R as a determination marker. It is more preferable to use IL-1β, IL-7, TNF-α, and sIL-6R in combination as a determination marker.
  • For a switch patient administered with tocilizumab (hereinafter, also referred to as a "tocilizumab therapy switch patient"), it is preferable to use at least one type selected from the group consisting of IL-1β, IL-5, IL-6, IL-7, IL-10, IL-12, IL-13, IL-15, FGFbasic, GM-CSF, IFN-γ, TNF-α, and VEGF as a determination marker.
  • For a naive patient administered with etanercept (hereinafter, also referred to as an "etanercept therapy naive patient"), it is preferable to use at least one type selected from the group consisting of IL-6, IP-10, IL-2, IL-13, IL-15, sIL-6R, and sTNFRI as a determination marker. It is more preferable to use a combination of IL-2, IL-15, sIL-6R, and sTNFRI as a determination marker.
  • <Cases where value of disease activity indicator after therapy itself is predicted and determined for biological formulation>
  • For a tocilizumab therapy naive patient, it is preferable to use at least one type selected from the group consisting of sgp130, IL-8, Eotaxin, IP-10, sTNFRI, sTNFRII, IL-6, and VEGF as a determination marker. It is more preferable to use a combination of sgp130, IL-8, Eotaxin, IP-10, sTNFRI, sTNFRII, and IL-6, or a combination of sgp130, IL-8, Eotaxin, IP-10, sTNFRI, sTNFRII, IL-6, and VEGF as a determination marker.
  • For a tocilizumab therapy switch patient, it is preferable to use at least one type selected from the group consisting of sgp130, IL-1β, IL-2, IL-5, IL-15, GM-CSF, IFN-γ, TNF-α, and IP-10 as a determination marker. It is more preferable to use a combination of sgp130, IP-10, and GM-CSF as a determination marker.
  • For an etanercept therapy naive patient, it is preferable to use at least one type selected from the group consisting of IL-9, IL-6, IL-13, TNF-α, and VEGF as a determination marker. It is more preferable to use a combination of IL-9, TNF-α, and VEGF, or a combination of IL-6 and IL-13 as a determination marker.
  • <Cases where possibility of remission (whether remission is reached) by therapy is predicted and determined for biological formulation>
  • For patients administered with tocilizumab (including both tocilizumab therapy naive patients and tocilizumab therapy switch patients), it is preferable to use at least one type selected from the group consisting of sgp130, IP-10, sTNFRII, IL-6, IL-7, MCP-1 and IL-1β as a determination marker. It is more preferable to use at least sgp130, and even more preferable to use a combination of (i) sgp130, (ii) IP-10, (iii) sTNFRII, and (iv) IL-6, IL-7, MCP-1 or IL-1β. More specifically, for tocilizumab therapy naive patients, a combination of (i) sgp130, (ii) IP-10, (iii) sTNFRII, and (iv) IL-6, IL-7, MCP-1 or IL-1β is especially preferable as a determination marker. Further, for tocilizumab therapy switch patients, a combination of (i) sgp130, (ii) IP-10, (iii) sTNFRII, and (iv) IL-6 or IL-1β is especially preferable as a determination marker.
  • For an etanercept therapy naive patient, it is preferable to use at least one type selected from the group consisting of IL-9, TNF-α, VEGF, PDGF-bb, and MIP-1α as a determination marker. It is especially preferable to use a combination of IL-9 and TNF-α, a combination of VEGF and PDGF-bb, or a combination of MIP-1α and PDGF-bb as a determination marker.
  • It is known that serum concentration of each of the cytokines, chemokines, and soluble receptors used as a determination marker can be measured by a measurement system utilizing an antigen-antibody reaction such as ELISA. Such measuring kits are commercially available. Thus , the cytokines , chemokines, and soluble receptors can be measured with a known measuring kit by a known method in the determining method of the present invention.
  • Prediction and determination of therapeutic effect due to biological formulation
  • A therapeutic effect due to a biological formulation can be predicted and determined based on a measured value of the determination marker. For example, the prediction and determination include a method in which the determination marker is measured in advance for patients in full remission and patients who are not in remission from therapy with a biological formulation; a regression equation of a measured value of the determination marker (explanatory variable) and a therapeutic effect of biological formulation (objective variable) are found by regression analysis; and a measured value of a determination marker of a rheumatoid arthritis patient targeted for determination is applied to said regression equation. When finding a regression equation, it is preferable to use a log value of serum concentration (pg/ml) for the determination markers other than sgp130. For sgp130, a value of serum concentration (µg/ml) is preferably used. Further, it is preferable that a regression equation is derived by multiple regression analysis. The objective variable in the above-described regression equation may be appropriately determined based on the therapeutic effect to be predicted and determined.
  • For example, when predicting and determining the level of improvement in a symptom after therapy for a biological formulation, the objective variable may be set to "a value obtained by subtracting a value of a disease activity indicator after a predetermined period of therapy from a value of a disease activity indicator prior to therapy" for analysis by multiple linear regression analysis. For example, when predicting and determining a value of a disease activity indicator after therapy for a biological formulation, the objective variable may be set to "a value of disease activity indicator after a predetermined period of therapy" for analysis by multiple linear regression analysis. In this regard, specific examples of a value of a disease activity indicator include a DAS (Disease activity score)-28 value, CDAI (Clinical Disease Activity Index) value, SDAI (Simple Disease Activity Index) value and the like. A DAS-28 value, CDAI value and SDAI value are correlated with one another and reflect a symptom of rheumatoid arthritis. Thus, any of such disease activity indicator values may be used in the determining method of the present invention. Further, a disease activity indicator used in the determining method of the present invention is not limited to those exemplified above. Indicators that may be newly advocated in the future can be used.
  • Further, when predicting or determining the possibility of remission due to therapy with a biological formulation (result of whether there is remission or no remission), multiple logistic regression analysis may be used for analysis.
  • For regression analysis utilizing a measured value of the determination marker as an explanatory variable, a value of a disease activity indicator prior to therapy (DAS-28 value, CDAI value, SDAI value or the like) or a result of evaluation by a Boolean method may be utilized as an explanatory variable.
  • Hereinafter, therapeutic effects to be predicted and determined are separated into a level of improvement in a symptom after therapy, DAS-28 value after therapy, and possibility of remission to disclose specific methods for the determining method of the present invention. However, the determining method of the present invention should not be interpreted to be limited to the following specific methods.
  • <Prediction and determination of level of improvement in symptom after therapy>
  • A level of improvement in a symptom after therapy due to a biological formulation can be predicted and determined by multiple linear regression analysis while setting an objective variable as "a value obtained by subtracting a value of a disease activity indicator after a predetermined period of therapy from a value of a disease activity indicator prior to therapy" and an explanatory variable as "a measured value of the determination marker".
  • In Examples described below, the following equations (1) and (2) have been discovered as regression equations for predicting and determining a level of improvement in a symptom after 16 weeks of therapy due to a biological formulation (level of improvement in DAS-28 value; DAS-28 value prior to therapy - DAS-28 value after 16 weeks of therapy), separated by the past dosing history of a rheumatoid arthritis patient and type of biological formulation. A level of improvement in a symptom after 16 weeks of therapy can be predicted and determined by finding an objective variable from applying values to one of the following regression equations (1) and (2) depending on the past dosing history of a rheumatoid arthritis patient subjected to determination and type of biological formulation. A level of improvement in a symptom due to a biological formulation is predicted and determined to be large for the patient for larger values of the objective variable calculated by the following regression equation.
  • [Cases where level of improvement in symptom after 16 weeks of therapy (level of improvement in DAS-28 value; DAS-28 value prior to therapy - DAS28-value after 16 weeks of therapy) is predicted and determined for tocilizumab therapy naïve patient]
  • Determination markers: IL-1β, IL-7, TNF-α, and sIL-6R Objective function DAS 28 value prior to the therapy DAS 28 value after 16 weeks of therapy = 5.505 + 3.618 × A + 3.255 × B + 1.475 × C + 1.841 × D
    Figure imgb0001
    1. A: log value of serum IL-1β concentration (pg/ml)
    2. B: log value of serum IL-7 concentration (pg/ml)
    3. C: log value of serum TNF-α concentration (pg/ml)
    4. D: log value of serum sIL-6R concentration (pg/ml)
    [Cases where level of improvement in symptom after 16 weeks of therapy (level of improvement in DAS-28 value; DAS-28 value prior to therapy - DAS28-value after 16 weeks of therapy) is predicted and determined for etanercept therapy naive patient]
  • Determination markers: IL-2, IL-15, sIL-6R, and sTNFRI Objective function DAS 28 value prior to the therapy DAS 28 value after 16 weeks of therapy = 7.325 + 1.567 × E + 1.632 × F + 2.540 × D + 1.973 × G
    Figure imgb0002
    • E: log value of serum IL-2 concentration (pg/ml)
    • F: log value of serum IL-15 concentration (pg/ml)
    • D: log value of serum sIL-6R concentration (pg/ml)
    • G: log value of serum sTNFRII concentration (pg/ml)
  • The regression equations (1) and (2) demonstrate an example of a regression equation used to predict and determine a level of improvement in DAS-28 value after 16 weeks of therapy due to a biological formulation. However, a level of improvement in a CDAI value or an SDAI value after 16 weeks of therapy due to a biological formulation (level of improvement in CDAI value or SDAI value; CDAI value or SDAI value prior to therapy - CDAI value or SDAI value after 16 weeks of therapy) can naturally be predicted and determined by multiple linear regression analysis by the same method using a CDAI value or SDAI value. Further, since a therapeutic effect stabilizes and appears after 16 weeks of therapy by a biological formulation, regression equations for predicting and determining a level of improvement in a symptom after 16 weeks of therapy are shown in the above-described regression equations (1) and (2). Naturally, a level of improvement in a symptom before or after 16 weeks of therapy due to the biological formulations can be predicted and determined by multiple linear regression analysis using the same method.
  • In Examples described below, the following equations (3)-(7) have been discovered as regression equations for predicting and determining a DAS-28 value of a symptom after 16 weeks of therapy due to a biological formulation, separated by the past dosing history of a rheumatism patient and type of biological formulation. A DAS-28 value after 16 weeks of therapy can be predicted and determined by finding an objective variable from applying values to one of the following regression equations (3)-(7), depending on the past dosing history of a rheumatoid arthritis patient subjected to determination and type of biological formulation. When the objective variable calculated by the following regression equation is 2.3 or less, the patient is predicted and determined to reach remission due to a biological formulation.
  • [Cases where DAS-28 value after 16 weeks of therapy is predicted and determined for tocilizumab therapy naive patient]
  • Determination markers: sgp130, IL-8, Eotaxin, IP-10, sTNFRI, sTNFRII, IL-6, and VEGF Objective function DAS 28 value after 16 weeks of therapy = 6.909 + 5.341 × H + 3.940 × I 1.039 × J + 1.002 × K + 2.580 × L + 1.407 × G + 0.744 × M + 0.850 × N
    Figure imgb0003
    • H: serum sgp130 concentration (µg/ml)
    • I: log value of serum IL-8 concentration (pg/ml)
    • J: log value of serum Eotaxin concentration (pg/ml)
    • K: log value of serum IP-10 concentration (pg/ml)
    • L: log value of serum sTNFRII concentration (pg/ml)
    • G: log value of serum sTNFRII concentration (pg/ml)
    • M: log value of serum IL-6 concentration (pg/ml)
    • N: log value of serum VEGF concentration (pg/ml)
    [Cases where DAS-28 value after 16 weeks of therapy is predicted and determined for tocilizumab therapy naive patient]
  • Determination markers: sgp130, IL-8, Eotaxin, IP-10, sTNFRI, sTNFRII, and IL-6 Objective function DAS 28 value after 16 weeks of therapy = 4.731 + 5.433 × H + 2.551 × I 0.937 × J + 1.116 × K + 2.010 × L + 1.630 × G + 0.577 × M
    Figure imgb0004
    • H: serum sgp130 concentration (µg/ml)
    • I: log value of serum IL-8 concentration (pg/ml)
    • J: log value of serum Eotaxin concentration (pg/ml)
    • K: log value of serum IP-10 concentration (pg/ml)
    • L: log value of serum sTNFRII concentration (pg/ml)
    • G: log value of serum sTNFRII concentration (pg/ml)
    • M: log value of serum IL-6 concentration (pg/ml)
    [Cases where DAS-28 value after 16 weeks of therapy is predicted and determined for tocilizumab therapy switch patient]
  • Determination markers: sgp130, IP-10, and GM-CSF Objective function DAS 28 value after 16 weeks of therapy = 2.837 + 6.037 × H + 0.714 × K + 0.622 × O
    Figure imgb0005
    • H: serum sgp130 concentration (µg/ml)
    • K: log value of serum IP-10 concentration (pg/ml)
    • O: log value of serum GM-CSF concentration (pg/ml)
    [Cases where DAS-28 value after 16 weeks of therapy is predicted and determined for etanercept therapy naive patient]
  • Determination markers: IL-6 and IL-13, DAS-28 value prior to etanercept administration is also used as an explanatory variable Objective function DAS 28 value after 16 weeks of therapy = 0.081 + 0.522 × a + 0.969 × M + 1.409 × P
    Figure imgb0006
    • a: DAS-28 value prior to etanercept administration
    • M: log value of serum IL-6 concentration (pg/ml)
    • P: log value of serum IL-13 concentration (pg/ml)
    [Cases where DAS-28 value after 16 weeks of therapy is predicted and determined for etanercept therapy naive patient]
  • Determination markers: IL-9, TNF-α and VEGF Objective function DAS 28 value after 16 weeks of therapy = 0.703 + 0.646 × S + 0.551 × C + 0.858 × N
    Figure imgb0007
    • S: log value of serum IL-9 concentration (pg/ml)
    • C: log value of serum TNF-α concentration (pg/ml)
    • N: log value of serum VEGF concentration (pg/ml)
  • Since regression equation (7) does not use a DAS-28 value prior to etanercept administration as an explanatory variable, a DAS-28 value after 16 weeks of therapy can be predicted while eliminating a subjective opinion of a physician. Thus, regression equation (7) is considered preferable over regression equation (6).
  • The regression equations (3)-(7) show examples of a regression equation used to predict and determine a DAS-28 value after 16 weeks of therapy due to a biological formulation. However, a CDAI value or SDAI value itself after 16 weeks of therapy due to a biological formulation can naturally be predicted and determined by multiple linear regression analysis by the same method using a CDAI value or SDAI value. Further, as discussed above, since a therapeutic effect stabilizes and appears after 16 weeks of therapy due to a biological formulation, regression equations for predicting and determining a value of a disease activity indicator after 16 weeks of therapy are shown in the above-described regression equations (3)-(7). However, a value of disease activity indicator prior to or after 16 weeks of therapy due to the biological formulations can naturally be predicted and determined by multiple linear regression analysis using the same method.
  • In Examples described below, the following equations (8)-(16) have been discovered as regression equations for predicting and determining the possibility of remission (either remission or not in remission) after 16 weeks of therapy due to a biological formulation, separated by the past dosing history of a rheumatoid arthritis patient and type of biological formulation. It is possible to predict and determine whether remission is reached after 16 weeks of therapy by finding the probability (p) of remission after 16 weeks of therapy from applying values to one of the following regression equations (8)-(16) depending on the past dosing history of a rheumatoid arthritis patient subjected to determination and type of biological formulation. The probability of remission estimated from the following regression equations (8) - (16) refers to the probability of a DAS-28 value being 2.3 or less. A p value computed from regression equations (8)-(16) closer to 1 indicates a higher possibility of remission after 16 weeks of therapy. For example, the p value of 0.5 or higher can predict and determine remission and less than 0.5 can predict and determine no remission for convenience's sake. In this regard, a DAS-28 value of 2.3 is used as the boundary between remission and non-remission to enhance the precision of prediction and determination of remission because a CRP value tends to decrease and DAS-28 value may decreases regardless of inflammation by inhibiting IL-6. The value is set at a lower value of DAS-28 value (2.6), which is generally considered the boundary between remission and non-remission.
  • [Cases where possibility of remission is predicted and determined for tocilizumab therapy naive patient]
  • Determination markers: sgp130, IP-10, sTNFRII, and IL-6 p / 1 p = exp 5.095 + 36.648 × H + 4.004 × K + 5.632 × G + 1.658 × M
    Figure imgb0008
    • p: probability of remission after 16 weeks of therapy H: serum sgp130 concentration (µg/ml)
    • K: log value of serum IP-10 concentration (pg/ml)
    • G: log value of serum sTNFRII concentration (pg/ml)
    • M: log value of serum IL-6 concentration (pg/ml)
    [Cases where possibility of remission is predicted and determined for tocilizumab therapy naive patient]
  • Determination markers: sgp130, IP-10, sTNFRII, and IL-7 p / 1 p = exp 3.467 + 42.849 × H + 4.430 × K + 5.736 × G + 2.705 × B
    Figure imgb0009
    • p: probability of remission after 16 weeks of therapy
    • H: serum sgp130 concentration (µg/ml)
    • K: log value of serum IP-10 concentration (pg/ml)
    • G: log value of serum sTNFRII concentration (pg/ml)
    • M: log value of serum IL-7 concentration (pg/ml)
    [Cases where possibility of remission is predicted and determined for tocilizumab therapy naive patient]
  • Determination markers: sgp130, IP-10, sTNFRII, and MCP-1 p / 1 p = exp 2.834 + 38.721 × H + 4.664 × K + 5.369 × G + 2.502 × B
    Figure imgb0010
    • p: probability of remission after 16 weeks of therapy
    • H: serum sgp130 concentration (µg/ml)
    • K: log value of serum IP-10 concentration (pg/ml)
    • G: log value of serum sTNFRII concentration (pg/ml)
    • P: log value of serum MCP-1 concentration (pg/ml)
    [Cases where possibility of remission is predicted and determined for tocilizumab therapy naive patient]
  • Determination markers: sgp130, IP-10, sTNFRII, and IL-1β p / 1 p = exp 1.269 + 39.538 × H + 3.807 × K + 5.086 × G + 1.647 × A
    Figure imgb0011
    • p: probability of remission after 16 weeks of therapy
    • H: serum sgp130 concentration (µg/ml)
    • K: log value of serum IP-10 concentration (pg/ml)
    • G: log value of serum sTNFRII concentration (pg/ml)
    • A: log value of serum IL-1β concentration (pg/ml)
    [Cases where possibility of remission is predicted and determined for tocilizumab therapy switch patient]
  • Determination markers: sgp130, IP-10, sTNFRII, and IL-6 p / 1 p = exp 10.935 + 29.051 × H + 4.466 × K + 2.067 × G + 2.757 × M
    Figure imgb0012
    • p: probability of remission after 16 weeks of therapy
    • H: serum sgp130 concentration (µg/ml)
    • K: log value of serum IP-10 concentration (pg/ml)
    • G: log value of serum sTNFRII concentration (pg/ml)
    • M: log value of serum IL-6 concentration (pg/ml)
    [Cases where possibility of remission is predicted and determined for tocilizumab therapy switch patient]
  • Determination markers: sgp130, IP-10, sTNFRII, and IL- p 1 p = exp 9.671 + 27.150 × H + 3.205 × K + 1.914 × G + 2.540 × A
    Figure imgb0013
    • p: probability of remission after 16 weeks of therapy
    • H: serum sgp130 concentration (µg/ml)
    • K: log value of serum IP-10 concentration (pg/ml)
    • G: log value of serum sTNFRII concentration (pg/ml)
    • A: log value of serum IL-1β concentration (pg/ml)
    [Cases where possibility of remission is predicted and determined for etanercept therapy naive patient]
  • Determination markers: VEGF and PDGF-bb, DAS-28 value prior to etanercept administration is also used as an explanatory variable. p / 1 p = exp 19.058 + 1.390 × a + 2.763 × E + 4.962 × Q
    Figure imgb0014
    • p: probability of remission after 16 weeks of therapy
    • a: DAS-28 value prior to etanercept administration
    • E: log value of serum VEGF concentration (pg/ml)
    • Q: log value of serum PDGF-bb concentration (pg/ml)
    [Cases where possibility of remission is predicted and determined for etanercept therapy naive patient]
  • Determination markers: MIP-1α and PDGF-bb, DAS-28 value prior to etanercept administration is also used as an explanatory variable. p / 1 p = exp 18.491 + 1.107 × a + 1.808 × R + 3.930 × Q
    Figure imgb0015
    • p: probability of remission after 16 weeks of therapy
    • a: DAS-28 value prior to etanercept administration
    • R: log value of serum MIP-1α concentration (pg/ml)
    • Q: log value of serum PDGF-bb concentration (pg/ml)
    [Cases where possibility of remission is predicted and determined for etanercept therapy naive patient]
  • Determination markers: IL-9 and TNF-α p / 1 p = exp 1.004 + 1.711 × S + 1.031 × C
    Figure imgb0016
    • p: probability of remission after 16 weeks of therapy
    • S: log value of serum IL-9 concentration (pg/ml)
    • C: log value of serum TNF-α concentration (pg/ml)
  • Since the regression equation (16) does not use a DAS-28 value prior to etanercept administration as an explanatory variable, the possibility of remission can be predicted while eliminating a subjective opinion of a physician. Thus, regression equation (16) is considered preferable over regression equations (14) and (15).
  • The regression equations (6)-(16) show examples of a regression equation for predicting and determining the possibility of remission after 16 weeks of therapy, with a DAS-28 value after 16 weeks of therapy of 2.3 or lower considered remission and the value over 2.3 as non-remission. However, the possibility of remission after 16 weeks of therapy due to a biological formulation can naturally be predicted and determined by multiple logistic regression analysis with the same method using a CDAI value or SDAI value. Further, as discussed above, since a therapeutic effect stabilizes and appears after 16 weeks of therapy by a biological formulation, regression equations for predicting and determining the possibility of remission after 16 weeks of therapy are shown in the above-described regression equations (6) - (16). However, the possibility of remission prior to or after 16 weeks of therapy due to a biological formulation can be predicted and determined by multiple logistic regression analysis using the same method.
  • Selection of biological formulation to be administered
  • The determining method of the present invention can predict the therapeutic effectiveness of a biological formulation prior to the administration thereof. Thus, the method can be utilized in selecting the optimal biological formulation that should be administered prior to starting therapy.
  • For example, for a level of improvement in a symptom after therapy of a naive patient, cases in which tocilizumab is administered and cases in which etanercept is administered are each predicted by the aforementioned method and a biological formulation with a higher level of improvement is selected, so that an optimal biological formulation can be administered to the patient. Specifically, a level of improvement in a symptom after therapy with tocilizumab therapy, which is predicted by using regression equation (1) is compared to a level of improvement in a symptom after therapy with etanercept therapy, which is predicted by using regression equation (2), so that the biological formulation with a higher level of improvement can be selected as the optimal biological formulation.
  • For example, for a DAS-28 value after 16 weeks of therapy of a naive patient, cases in which tocilizumab is administered and cases in which etanercept is administered are each predicted by the aforementioned method and a biological formulation with a lower DAS-28 value after 16 weeks of therapy is selected so that an optimal biological formulation can be administered to the patient. Specifically, a DAS-28 value after 16 weeks of therapy with tocilizumab therapy, which is predicted by using one of regression equations (3)-(5) is compared to a DAS-28 value after 16 weeks of therapy with etanercept therapy, which is predicted by using regression equation (6) or (7), so that the biological formulation with a smaller DAS-28 value can be selected as the optimal biological formulation.
  • For example, for the possibility of remission of a naive patient, cases in which tocilizumab is administered and cases in which etanercept is administered are each predicted by the aforementioned method to select a biological formulation with a higher possibility of remission, so that the optimal biological formulation can be administered to the patient. Specifically, the possibility of remission with tocilizumab therapy, which is predicted by using one of regression equations (8) - (11), is compared to the possibility of remission , which is predicted by using one of regression equations (14)-(16), so that the biological formulation with a higher possibility of remission can be selected as the optimal biological formulation.
  • 2. Diagnostic agent
  • The present invention further provides a diagnostic agent for carrying out the above-described detection method. Specifically, the diagnostic agent of the present invention is a diagnostic agent for determining the effectiveness of therapy due to a biological formulation targeting an inflammatory cytokine for a rheumatoid arthritis patient, characterized by comprising a reagent capable of detecting at least one type of determination marker selected from the group consisting of sgp130, IP-10, sTNFRI, sTNFRII, GM-CSF, IL-1β, IL-2, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12, IL-13, IL-15, Eotaxin, VEGF, MCP-1, TNF-α, IFN-γ, FGFbasic, PDGF-bb, sIL-6R, and MIP-1α.
  • The determination marker can be measured by a measurement system utilizing an antigen-antibody reaction such as ELISA. Specific examples of reagents capable of detecting the determination marker include antibodies that can specifically bind to the determination marker and fragments thereof. Further, antibodies that can specifically bind to the determination marker may be bound on a suitable support to be provided as an antibody array.
  • Furthermore, the diagnostic agent of the present invention may comprise a reagent (secondary antibody, color producing substance or the like) required for detecting the determination marker by an antigen-antibody reaction.
  • Examples
  • Hereinafter, the present invention is disclosed in detail while using Examples. However, the present invention is not limited thereby.
  • 1. Patient and Experimental Method (Patient)
  • Hereinafter, a rheumatism patient who has not received anti-cytokine therapy (administration of infliximab, etanercept, adalimumab, tocilizumab or the like) in the past is referred to as a naive patient, and a rheumatism patient who has received anti-cytokine therapy in the past is referred to as a switch patient.
  • 155 rheumatoid arthritis patients, to whom methotrexate therapy was ineffective, were registered at the Higashihiroshima Memorial Hospital from March 2008 to June 2013. Among the 155 patients, 98 patients received therapy with tocilizumab and the remaining 57 patients received therapy with etanercept. Among the 98 patients who received therapy with tocilizumab, 58 patients were naive patients who had not previously received anti-cytokine therapy and 40 patients were switch patients who had previously received anti-cytokine therapy 1-3 times. Among 57 patients who received etanercept therapy, 49 patients were naive patients who had not previously received anti-cytokine therapy, and the remaining 8 patients were switch patients who had previously received anti-cytokine therapy. Informed consent was obtained prior to receiving a blood sample supply from all patients. Further, the tests were conducted with permission prior to the study from the ethics committee of the Higashihiroshima Memorial Hospital.
  • Table 1 shows the clinical baseline individual group statistics for group of individuals and clinical diagnosis. Further, Figure 1 shows a trial profile of patients receiving therapy with tocilizumab and patients receiving therapy with etanercept. Figures 2-1 to 2-4 show serum concentration of cytokine/chemokine/soluble receptor prior to therapy. 9 naïve patients (8 patients who suffered from side effects or other diseases and 1 patient for whom data could not be obtained for the entire 16 weeks) among patients treated with tocilizumab were eliminated. Further, 1 switch patient suffering from a side effect (patient for whom data could not be obtained for the entire 16 weeks) among patients treated with tocilizumab was eliminated. There was hardly any difference in DAS-28 value, CRP, swollen joint count, tender joint count, Stage, and Class among the groups (Table 1). Further, the duration of disease was shorter for patients treated with etanercept in comparison to patients treated with tocilizumab (Table 1).
  • In order to create a baseline concentration of cytokines, serum was collected from healthy individuals (56 individual; 20 males and 36 females) without a history of suffering from hepatitis C or cancer. The healthy individuals underwent medical examination by the Louis Pasteur Center for Medical Research or the Higashihiroshima Memorial Hospital and informed consent was received in writing from the healthy individuals. The baseline concentration was used to find a distribution pattern of cytokines/chemokines/soluble receptors.
  • (Experimental Method)
  • Prior to therapy, concentrations of cytokines, chemokines, and soluble receptors in the serum of rheumatoid arthritis patients were measured.
  • Figure 1 shows clinical results for naive patients and switch patients administered with 8 mg/kg tocilizumab or 50 mg/kg etanercept once every 4 weeks. After 16 weeks of therapy (after 4 administrations), a therapeutic effect was determined based on DAS-28-CRP values and whether the patient is in remission or non-remission. Results for non-remission were further classified into low, medium and high based on DAS-28-CRP values of the patients. DAS-28-ESR values are extensively used to determine the symptom of rheumatoid arthritis patients. However, it is reported that DAS-28-CRP values are almost interchangeable with DAS-28-ESR values and the same results are derived therefrom (Ann Rheum Dis. 2007, March 407-409 Comparison of Disease Activity Score (DAS)28-erythrocyte Sedimentation rate and DAS-C-reactive protein threshold votes. Inoue E, Yamanaka H, et al.)
  • In the present tests, DAS-28-CRP values were used to determine the symptoms of rheumatoid arthritis patients. Remission was classified as DAS-28-CRP value < 2.3 and non-remission was classified as DAS-28-CRP value ≥ 2.3. Furthermore, DAS-28-CRP classification system developed by Inoue et al was used to classify non-remission patients as low (DAS-28-CRP value = 2.3-2.6), medium (DAS-28-CRP value = 2.7-4.1) and high (DAS-28-CRP value > 4.1) depending of the severity of the symptoms. To obtain consistent determination of symptoms, the same physician at the Higashihiroshima Memorial Hospital determined the final symptoms of all patients. Further, Figure 3 shows detailed clinical results of each patient shown in Figure 1, i.e., results of determining DAS-28-CRP values prior to therapy and after 16 weeks of therapy for naive patients who received tocilizumab therapy, switch patients who received tocilizumab therapy, and naive patients who received etanercept therapy. Hereinafter, DAS-28-CRP values may be denoted simply as DAS-28 values.
  • (Analysis of cytokine/chemokine/soluble receptor)
  • For all measurements of cytokines, a multiplex cytokine array system (Bio-Plex 200, Bio-Rad Laboratories) was used in accordance with the product protocol thereof. Serum for all patients and healthy individuals were collected by 10 minutes of centrifugation (1600xg). All serum samples were stored at -80° C. Bio-Plex Human Cytokine 27-Plex Panel is configured such that 27 types of cytokines (IL-1β, IL-1RA, IL-2, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12 (p70), IL-13, IL-15, IL-17, basic FGF, eotaxin, G-CSF, GM-CSF, IFN-γ, IP-10, MCP-1, MIP-1α, MIP-1β, PDGF-bb, RANTES, TNF-α, and VEGF) can be analyzed. In addition thereto, sIL-6R, sgp130, sTNF-RI and sTNF-RII were also analyzed (Milliplex®MAP, Human Soluble Cytokine Receptor Panel: Millipore Co. MA). In the present test, concentrations of cytokines, chemokines, and soluble receptors of 56 healthy individuals were simultaneously measured to find the distribution patterns thereof. Bio-Plex Manager software version 5.0 was used to conduct data collection and analysis.
  • (Statistical Analysis)
  • The distribution of cytokine/chemokine values in healthy individuals was analyzed. The log values of the values of the concentration (pg/ml) of cytokines, chemokines, and soluble receptors, other than sgp130, were used for the analysis. The values of concentration (µg/ml) were directly used for sgp130.
  • First, simple linear regression analysis and multiple linear regression analysis were performed to investigate the association between cytokine/chemokine/soluble receptor concentration or clinical test values and values obtained by subtracting DAS-28 values after 16 weeks from DAS-28 values of patients at 0 weeks. Next, DAS-28 values after 16 weeks were estimated from a value computed from regression introduced with the clinical test values. Furthermore, simple logistic regression analysis and multiple logistic regression analysis were preformed to analyze the relationship between serum cytokine concentration and remission or non-remission. The resulting parameter p value of <0.05 indicates the presence of a significant difference. All statistical analysis was conducted by using the JMP 9.0 software.
  • 2. Results (Clinical evaluation)
  • Tables 1 and 2 show clinical baseline individual group statistics, clinical diagnosis and cytokine/chemokine/soluble receptor characteristics. Figures 2-1 to 2-4 shows clinical baseline individual group statistics for healthy individuals and rheumatoid arthritis patients with respect to serum concentrations of cytokines/chemokines/soluble receptors prior to therapy. In Table 1, Stage and Class are results of determination with respect to functional classification criteria for rheumatoid arthritis based on Steinbrocker (1949) classification (I-IV; Steinbrocker O et al: Therapeutic criteria in rheumatoid arthritis. JAMA 140:659,1949) and Hochberg (1992) classification (I-IV; Hochberg MC et al, The American College of Rheumatology 1991 revised criteria for the Classification of global functional status in rheumatoid arthritis. Arthritis and Rheumatism, 35:498-502, 1992), respectively. The results indicate that there is no clinically significant difference among the three rheumatism patient groups and the serum concentrations of cytokines/chemokines/soluble receptors in most rheumatism patients are significantly higher in comparison to healthy individuals.
    Figure imgb0017
    Figure imgb0018
    Figure imgb0019
  • 49 naive patients received 16 weeks of tocilizumab therapy. 56% of the patients thereof (27 patients) exhibited remission and the rest of the 21 patients exhibited non-remission (Figure 1). Furthermore, 39 switch patients received 16 weeks of tocilizumab therapy, of which 9 patients exhibited remission and the rest of the 30 patients exhibited non-remission.
  • Further, 49 naive patients received 16 weeks of etanercept therapy. 18 patients thereamong exhibited remission and the remaining 31 patients exhibited non-remission (Figure 1). In the present test, the number of switch patients was extremely low. Thus, only naive patients were used in the analysis for etanercept therapy.
  • Figure 4 shows the relationship between a DAS-28 value prior to therapy and a value obtained from subtracting a DAS-28 value after 16 weeks from the DAS-28 value prior to therapy (PreDAS-28score - 16W DAS-28 score) in a naive patient who has received tocilizumab therapy (PreDAS-28 score). According to Figure 4, improvement in DAS-28 values was observed after tocilizumab therapy in most naive patients.
  • (Search for biomarkers based on DAS-28 value after 16 weeks of therapy by using serum concentration of cytokines/chemokines/soluble receptors in rheumatism patient prior to therapy)
  • According to the current results, about 55% of naive patients and about 23% of switch patients are expected to exhibit remission after tocilizumab therapy. Although the final symptoms are not identical, some improvement in the symptom is observed in about 45% of naive patients and about 77% of switch patients. Further, about 36.7% of naive patients are expected to exhibit remission after etanercept therapy. In addition, some improvement in the symptom is observed after etanercept therapy in about 60% of the remaining patients.
  • It has been reported by Pers Y.M. et al (Rheumatology (2013) doi: 10.1093/rheumatology/ket301 First published online: September 19, 2013) that when DAS-28 values after 12-24 weeks of tocilizumab therapy were assessed from general medical examination, 40% of patients exhibited remission, and the main prediction markers were young patients, high CRP value, and patients without a cardiovascular disorder. Further, Koike T (J. Rheumatology, November 2013) et al reported 47.6% remission with respect to DAS28-ESR after 28 weeks of tocilizumab therapy. Meanwhile for etanercept, it is reported by Markenson JA et al (J. Rheumatology, July 2011, p. 1273-81) in the RADIUS study and Curitis JR et al (Ann RheumDis. 2012. 71. 206-212) in the TEMPO study that patients achieving a low disease activity of DAS-28 value ≤ 3.2 after 52 weeks and remission were 53%, and 63% when methotrexate is added. Further, Koike T et al (J. Rheumatology, October 2013, p. 1658-1668) have demonstrated that therapy of etanercept adding with methotrexate, when assessing DAS-28 values after 24 weeks, was more effective than therapy with etanercept alone or therapy adding an anti-rheumatism agent (DMARD) other than methotrexate. Furthermore, it is reported by Cannon GW et al (Clin Exp Rheumatol. November 2013) that 35% reached remission in a 3 year observation from TEMPO and RADIUS studies. Furthermore, Cannon GW et al demonstrated that patients with low disease activity are more likely exhibit remission. However, these reports do not reveal a marker for predicting and determining a therapeutic effect due to a biological formulation.
  • In this regard, serum concentrations of cytokines/chemokines were used to investigate whether it is possible to estimate the level of improvement in rheumatoid arthritis based on DAS-28 values after 16 weeks of therapy.
  • Figures 2-1 to 2-4 show comparisons of baseline individual group statistics of cytokines/chemokines/soluble receptors prior to therapy for healthy individuals and three groups (naive patients who received tocilizumab therapy, switch patients who received tocilizumab therapy, naive patients who received etanercept therapy). As can be seen from Figures 2-1 to 2-4, serum concentration other than those for sgp130, sIL-6R and sTNFRI in rheumatoid arthritis patients were significantly higher in comparison to healthy individuals. Further, serum concentrations of cytokines/chemokines were lower in naive patients who received etanercept therapy in comparison to naive patients who received tocilizumab therapy. Further, it was revealed from this result that naive patients who received tocilizumab therapy have a higher CRP value prior to therapy in comparison to naive patients who received etanercept therapy.
  • Simple linear regression analysis was performed to find the cytokine/chemokine involved in the level of improvement in DAS-28 values. The level of improvement in a DAS-28 value (DAS-28 value prior to therapy - DAS-28 value after 16 weeks of therapy) was used as an objective variable and serum concentration of cytokines/chemokines/soluble receptors were used directly, or by converting into a log value, as an independent variable. The results are shown in Table 2. As shown in Table 2, logIL-7, logIL-8, logIL-12, logIL-13, logIP-10 and logVEGF exhibiting p < 0.05 significantly matched the level of improvement in DAS-28 values in naive patients who received tocilizumab therapy. Further, for switch patients who received tocilizumab therapy, logIL-1β, logIL-5, logIL-6, logIL-7, logIL-10, logIL-12, logIL-13, logIL-15, logFGF, logGM-CSF, logIFN-γ, logTNF-α and logVEGF significantly matched the level of improvement in DAS-28 values. Meanwhile, logIL-6 and logIP-10 significantly matched the level of improvement in DAS-28 values for naive patients who receivedetanercept therapy.
  • Simple linear regression analysis Level of improvement in DAS-28
  • Figure imgb0020
  • Multiple linear regression analysis was performed to find the correlation between the level of improvement in DAS-28 value and cytokine/chemokine/soluble receptor concentration. As a result, it was found by phased multiple regression analysis that a combination of logIL-1β, logIL-7, logTNF-α and logsIL-6R is significantly correlated with the level of improvement in DAS-28 values in naive patients who received tocilizumab therapy (Table 3).
  • Meanwhile, a combination of logIL-2, logIL-15, logIL-6R, and logTNFRI was found to have significant correlation with the level of improvement in DAS-28 values in naive patients who received etanercept therapy (Table 4). [Table 3]
    Multiple linear regression analysis on naïve patients who received tocilizumab therapy
    Level of improvement in DAS-28
    Objective variable: DAS-28 improvement
    (=0 week DAS-28 value - 16 week DAS-28 value)
    Naïve patients who received tocilizumab therapy
    Multiple regression analysis (Objective value=0w-16wDAS28)
    R^2 0.376
    ANOVA(Analysis of variance) p=0.0004
    Cytokine/Chemokine/soluble receptor Estimate p value
    intercept 5.505 0.1216
    logHu IL-1b -3.618 0.0002
    logHu IL-7 3.255 0.0002
    logHu TNF-α 1.475 0.0221
    logHu-sIL-6R -1.814 0.0264
    [Table 4]
    Naïve patients who received etanercept therapy
    Multiple linear regression analysis level of improvement in DAS-28
    (Objective variable : DAS-28 improvement (=0 week DAS-28 value - 16 week DAS-28 value)
    Naïve patients who received etanercept therapy
    Multiple regression analysis (Objective value=0w-16wDAS28)
    R^2 0.343
    ANOVA(Analysis of variance) p=0.0037
    Cytokine/Chemokine/sol uble receptor Estimate p value
    intercept 7.325 0.0231
    logHu IL-2 -1.567 0.0058
    logHu IL-15 1.632 0.0008
    logHusIL-6R -2.540 0.0130
    logHu-sTNFRI 1.973 0.0115
  • Simple linear regression analysis was performed to find the cytokine/chemokine/soluble receptor involved in the final assessment of a DAS-28 value after 16 weeks of therapy (16wDAS28). The DAS-28 value after 16 weeks of therapy was used as an objective variable and serum concentration of cytokines/chemokines/soluble receptors were used directly, or by converting into a log value, as an independent variable. As shown in Table 5, sgp130 exhibiting p < 0.05 significantly matched DAS-28 values after 16 weeks of therapy in naive patients who received tocilizumab therapy. Further, for switch patients who received tocilizumab therapy, logIL-1β, logIL-2, logIL-5, logIL-15, logGM-CSF, logIFN-γ, logTNF-α and sgp130 significantly matched DAS-28 values after 16 weeks of therapy. Meanwhile, logIL-9 significantly matched DAS-28 values after 16 weeks of therapy for naïve patients who received etanercept therapy. [Table 5]
    Simple linear regression analysis 16-week DAS-28
    Objective variable: 16-week DAS-28
    Simple linear repression enlysis of cytokine / chemokine / soluble receptor based on DAS-28 16w
    Simple linear regression anlysis were performed to find
    the parameters related to 16wDAS-28 (=16wDAS28).
    Naïve Tocilizumab Therapy Switch Tocilizumab Therapy Naïve Etanercept Therapy
    Tocilizumab naïve Tocilizumab switch Etanercept naïve
    Cytokine/Chemokine Estimates p value Estimates p value Estimates p value
    logHu IL-1b pg/ml 0.094 0.681 -0.604 0.035 -0.047 0.860
    logHu IL-1ra pg/ml -0.178 0.269 0.041 0.850 0.053 0.817
    logHu IL-2 pg/ml -0.078 0.644 -0.482 0.012 0.179 0.341
    logHu IL-4 pg/ml 0.335 0.426 -0.798 0.140 -0.190 0.661
    logHu IL-5 pg/ml -0.131 0.606 -0.832 0.025 0.119 0.724
    logHu IL-6 pg/ml 0.286 0.156 -0.301 0.216 0.095 0.712
    logHu IL-7 pg/ml 0.026 0.933 -0.617 0.119 0.199 0.550
    logHu IL-8 pg/ml 0.568 0.319 0.168 0.721 -0.175 0.756
    logHu IL-9 pg/ml -0.174 0.291 -0.190 0.330 0.545 0.011
    logHu IL-10 pg/ml -0.232 0.298 -0.395 0.163 0.351 0.217
    logHu IL-12 pg/ml -0.202 0.438 -0.529 0.115 0.413 0.177
    logHu IL-13 pg/ml -0.100 0.699 -0.533 0.096 0.467 0.236
    logHu IL-15 pg/ml -0.053 0.660 -0.325 0.019 0.092 0.557
    logHu IL-17 pg/ml -0.578 0.158 -0.819 0.262 -0.578 0.556
    logHu Eotaxin pg/ml -0.363 0.124 -0.360 0.291 0.056 0.868
    logHu FGF basic pg/ml -0.168 0.546 -0.688 0.085 0.493 0.281
    logHu G-CSF pg/ml -0.321 0.380 -0.978 0.120 -0.032 0.943
    logHu GM-CSF pg/ml -0.036 0.839 -0.589 0.001 0.191 0.368
    logHu IFN-g pg/ml 0.038 0.879 -0.709 0.024 0.015 0.960
    logHu IP-10 pg/ml -0.048 0.877 0.241 0.568 0.104 0.818
    logHu MCP-1 pg/ml 0.144 0.623 -0.113 0.739 0.009 0.981
    logHu MIP-1a pg/ml 0.196 0.591 -0.388 0.301 0.051 0.890
    logHu PDGF-bb pg/ml 0.097 0.798 -0.165 0.720 0.794 0.301
    logHu MIP-1b pg/ml 0.351 0.477 -0-284 0.573 -0.396 0.281
    logHu RANTES pg/ml 0.249 0.444 -0.452 0.224 0.382 0.631
    logHu TNF-a pg/ml -0.033 0.865 -0.646 0.012 0.035 0.875
    logHu VEGF pg/ml 0.400 0.136 -0.042 0.902 0.573 0.132
    sgp130 µ g/ml -3.785 0.046 -1.801 0.001 -3.005 0.207
    logHu-sIL-6R pg/ml -0.888 0.187 -1.246 0.075 -0.754 0.336
    logHu-sTNFRI pg/ml -1.028 0.039 0.033 0.955 0.094 0.902
    logHu-sTNFRII pg/ml -0.179 0.766 0.078 0.728 0.690 0.115
    DAS-28 0w 0.306 0.000 0.259 0.097 0.403 0.003
    MMP 0.000 0.645 0.000 0.464 0.002 0.023
    RF 0.001 0.378 0.000 0.818 0.000 0.504
    VAS 0.003 0.642 0.004 0.560 0.006 0.334
    Swollen joint count 0.069 0.000 0.066 0.183 0.081 0.022
    Tender joint count 0.067 0.000 0.074 0.166 0.042 0.193
    Stage 0.092 0.418 0.393 0.162 -0.197 0.232
    Class 0.188 0.483 0.130 0.680 0.453 0.096
  • Multiple linear regression analysis was performed to find the correlation between DAS-28 value after 16 weeks of therapy and cytokine/chemokine/soluble receptor concentration. As a result thereof, it was found by phased multiple regression analysis that a combination of sgp130, logIL-8, logEotaxin, logIP-10, logTNFRI, logTNFRII, logIL-6, and logIL-VEGF is significantly correlated with a DAS-28 value after 16 weeks of therapy in naive patients who received tocilizumab therapy as shown in Table 6. Further, it was found that there is a very significant correlation even without using logIL-VEGF (Table 7).
  • Further, it was found that a combination of sgp130, logIP-10, and logGM-CSF is significantly correlated with a DAS-28 value after 16 weeks of therapy in switch patients who received tocilizumab therapy (Table 8).
  • Meanwhile, a combination of DAS-28 value prior to therapy, logIL-6 and logIL-13 was also found to be significantly correlated with the level of improvement in DAS-28 value for naive patients who received etanercept therapy (Table 9). Further, a combination of logIL-9, logTNF-α, and logVEGF, even without using a DAS-28 value prior to therapy, is significantly correlated with a DAS-28 value after 16 weeks of therapy naive patients who received etanercept therapy (Table 10). [Table 6]
    Tocilizumab naïve multiple linear regression analysis
    Objective variable 16-week DAS-28
    Multiple linear regression anlysis of cytokine / chemokine / soluble receptor based on 16w DAS-28
    A.Multiple regression anlysis were performed to find the parameters related to 16wDAS-28 (=16wDAS28).
    Naïve Tocilizumab Therapy Tocilizumab naïve
    Multiple regression analysis (Objective value=16wDAS28)
    R^2 0.646
    ANOVA(Analysis of variance) P<0.0001
    Cytokine/Chemokine/soluble receptor Estimate p value
    mtercept 6.909 0.001
    sgp130# -0.534 0.002
    log IL-8 3.940 <.0001
    log Eotaxin -1.039 <.0001
    log IP-10 -1.002 0.002
    log sTNFRI -2.580 <.0001
    log sTNFRII 1.407 0.030
    log IL-6 0.744 0.002
    log VEGF -0.850 0.039
    sgp13o# : µ g/ml
    others : pg/ml
    [Table 7]
    Tocilizumab naïve multiple linear regression analysis
    Objective variable 16-week DAS-28
    Multiple linear regression anlysis of cytokine / chemokine / soluble receptor based on 16w DAS-28
    A.Multiple regression analysis were performed to find the parameters related to 16wDAS-28 (=16wDAS28).
    Naïve Tocilizumab Therapy Tocilizumab naïve
    Multiply regression analysis (objective value=16wDAS28)
    R^2 0.605
    ANOVA(Analysis of variance) p<0.0001
    Cytokine/Chemokine/soluble receptor Estimate p value
    intercept 4.731 0.0127
    sgp130# -0.543 0.003
    log IL-8 2.551 <.0001
    log Eotaxin -0.937 0.0004
    lop IP-10 -1.116 0.0007
    lop sTNFRI -2.010 0.0004
    log sTNFRII 1.630 0.0152
    log IL-6* 0.577 0.0096
    sgp130# : µ g/ml
    others : pg/ml
    [Table 8]
    Tocilizumab switch multiple linear regression analysis
    Objective variable 16-week DAS-28
    Multiple linear regression analysis of cytokine /chemokine /soluble receptor based on 16w DAS-28
    A Multiple regression anlysis were performed to find the parameters related to 16wDAS-28 (=16wDAS28).
    Tocilizumab switch
    Multiple regression analysis (Objective value=16wDAS28)
    R^2 0.486
    ANOVA(Analysis of variance) p<0.0001
    Cytokine/Chemokine/soluble receptor Estimate p value
    intercept 2.837 0.011
    sgp130# -0.604 0.003
    log IP-10 0.714 0.003
    log GM-CSF -0.622 0.0003
    sgp130# : µg/ml
    others : pg/ml
    [Table 9]
    Multiple linear regression analysis on naïve patients who received etanercept therapy
    Multiple linear regression analysis of cytokine/chemokine/soluble receptor and DAS28-CRP before therapy on 16week Das-28,
    Objective variable: 16-week DAS-28
    Naïve Etanercept Therapy
    Multiple regression analysis (Objective value:16wDAS28)
    R^2 0.321
    ANOVA(Analysis sof variance) p=0.0016
    Cytokine/Chemokine/soluble receptor estimate p value
    intercept 0.081 0.907
    DAS28-CRP (Prior to therapy) 0.522 0.000
    logHu IL-6 -0.969 0.015
    log HuIL-13 1.409 0.015
    [Table 10]
    Etanercept naïve multiple linear regression analysis.
    Objective variable 16-week DAS-28
    Multiple linear regression analysis of cytokine /chemokine /soluble receptor based on 16w DAS-28
    A Multiple regression analysis were performed to find the parameters related to 16wDAS-28 (=16wDAS28).
    Tocilizumab switch
    Multiple regression analysis (Objective value=16wDAS28)
    R^2 0.264
    ANOVA(Analysis of variance) p=0.0093
    Cytokine/Chemokine/soluble receptor Estimate p value
    intercept 0.703 0.348
    log IL-9 0.646 0.007
    log TNF-α -0.551 0.039
    log VEGF 0.858 0.053
    IL-9, TNF-α, VEGF : pg/ml
  • Further, regression equation (4) found based on the multiple linear regression analysis shown in Table 7 was used to find a predicted value of DAS-28 value after 16 weeks of therapy in naive patients who received tocilizumab therapy. Figure 5 shows the results of comparing predicted values of DAS-28 values after 16 weeks of therapy calculated by regression equation (4) and actual values of DAS-28 values after 16 weeks of therapy. It was confirmed from the results that DAS-28 values after 16 weeks of therapy estimated from the results of multiple linear regression analysis shown in Table 7 are very consistent with actual values of DAS-28 values after 16 weeks of therapy.
  • Further, regression equation (5) found based on the multiple linear regression analysis shown in Table 8 was used to find a predicted value of DAS-28 value after 16 weeks of therapy in switch patients who received tocilizumab therapy. Figure 6 shows the results of comparing the predicted values of DAS-28 values after 16 weeks of therapy calculated by regression equation (5) and actual values of DAS-28 values after 16 weeks of therapy. It was confirmed from the results that DAS-28 values after 16 weeks of therapy estimated from the results of multiple linear regression analysis shown in Table 8 are very consistent with actual values of DAS-28 values after 16 weeks of therapy.
  • Regression equation (7) found based on the multiple linear regression analysis shown in Table 10 was used to find a predicted value of DAS-28 value after 16 weeks of therapy in naive patients who received etanercept therapy. Figure 7 shows the results of comparing predicted values of DAS-28 values after 16 weeks of therapy calculated by regression equation (7) and actual values of DAS-28 values after 16 weeks of therapy. It was confirmed from the results that DAS-28 values after 16 weeks of therapy can be estimated to a certain extent from the results of multiple linear regression analysis shown in Table 10.
  • Further, a predicted value of DAS-28 after 16 weeks of therapy was found by using the aforementioned regression equation (4) while assuming that naive patients who received etanercept therapy had received tocilizumab therapy without receiving etanercept therapy. Figure 8 shows the actual values of DAS-28 after 16 weeks of etanercept therapy and predicted values of DAS-28 values after 16 weeks of therapy while assuming that tocilizumab therapy was received. From this result, naive patients who received etanercept therapy are classified into patients who are predicted to have a higher therapeutic effect when receiving tocilizumab therapy (Figure 8 a), patients who are predicted to have barely any difference observed between etanercept therapy and tocilizumab therapy (Figure 8 b), and patients who are predicted to have a higher therapeutic effect observed when receiving etanercept therapy (Figure 8 c). For patients shown in Figure 8 a, tocilizumab therapy is estimated to be more effective than etanercept therapy that was actually received. Thus, it was found that a more effective therapeutic agent can be selected by estimating DAS-28 values due to tocilizumab therapy and etanercept therapy prior to therapy by the present invention.
  • (Search for biomarkers for predicting and determining the possibility of remission by using serum concentration of cytokines/chemokines/soluble receptors in rheumatism patient prior to therapy)
  • In therapy of rheumatoid arthritis, it is desirable that even a partial improvement is observed in the symptom of a patient. However, it is most desirable to reach complete remission. In this regard, in addition to a search for various factors for estimating the final DAS-28 value, a search was conducted for cytokines/chemokines/soluble receptors for predicting whether a patient reaches complete remission.
  • Data for cytokine/chemokine/soluble receptor concentrations was analyzed for complete remission and non-remission patient groups by simple logistic regression analysis. Further, Table 10 shows the results of analyzing data for cytokine/chemokine/soluble receptor concentrations for naive patients and switch patients who received tocilizumab therapy and naive patients who received etanercept therapy. It was found by simple logistic regression analysis that swollen joint count and tender joint count and DAS-28 values were significantly different between complete remission and non-remission groups. Furthermore, sgp130 was significantly different between complete remission and non-remission groups in naive and switch patients who received tocilizumab therapy (Table 11) . Meanwhile, significant difference in sgp130 was not observed between remission and non-remission groups in naive patients who received etanercept therapy (Table 11). Further, Figure 9 shows the results of analyzing the relationship between serum sgp130 concentration and DAS-28 value prior to therapy for remission and non-remission patients. As is clear from Figure 9, many patients who have reached remission had a high sgp130 concentration. [Table 11]
    Simple logistic regression analysis
    Naïve patients who received tocilizumab therapy (n=48) Switch patients who received tocilizum ab therapy (n=40) Naïve patients who received etanercept therapy (n=43)
    Whole Model Test Parameter Estimates Whole Model Test Parameter Estimates Whole Model Test Parameter Estimates
    Cytokine/Chemokine Single logistic analysis, p value Estimates Single logistic analysis, p value Estimates Single logistic analysis, p value Estimates
    logHu IL-1b pg/ml 0.378 0.486 0.147 -1.081 0.577 -0.274
    logHu IL-1ra pg/ml 0.148 -0.628 0.087 1.573 0.323 0.478
    logHu IL-2 pg/ml 0.856 0.074 0.080 -1.009 0.857 0.064
    logHu IL-4 pg/ml 0.534 0.638 0.420 -1.241 0.965 -0.035
    logHu IL-5 pg/ml 0.814 0.143 0.189 -1.276 0.896 -0.083
    logHu IL-6 pg/ml 0.184 0.663 0.270 -0.710 0.950 -0.030
    logHu IL-7 pg/ml 0.585 0.401 0.233 -1.231 0.660 0.280
    logHu IL-8 pg/ml 0.432 1.088 0.864 -0.207 0.751 -0.333
    loghu IL-9 pq/ml 0.545 -0.242 0.289 -0.540 0.020 1.075
    loghu IL-10 pg/ml 0.604 -0.281 0.196 -0.948 0.572 0.310
    logHu IL-12 pg/ml 0.773 -0.181 0.113 -1.457 0.833 0.123
    logHu IL-13 pg/ml 0.963 0.029 0.432 -0.669 0.671 0.322
    logHu IL-15 pg/ml 0.924 0.027 0.173 -0.519 0.942 0.021
    logHu IL-17 pg/ml 0.197 -1.332 0.920 0.147 0.691 -0.730
    logHu Eotaxin pg/ml 0.447 -0.441 0.512 -0.590 0.639 0.299
    logHu FGF basic pg/ml 0.792 -0.177 0.725 -0.371 0.402 0.773
    logHu G-CSF pg/ml 0.599 -0.471 0.786 -0.450 0.901 -0.104
    logHu GM-CSF pg/ml 0.910 0.104 0.099 -0.930 0.798 -0.102
    logHu IFN-g pg/ml 0.536 0.369 0.190 -1.081 0.681 -0.228
    loghu IP-10 pg/ml 0.647 -0.344 0.604 0.557 0.393 0.733
    logHu MCP-1 pg/ml 0.402 0.593 0,696 -0.366 0.960 0.035
    logHu MIP-1a pg/ml 0.428 0.698 0.305 -0.963 0.885 -0.098
    logHu PDGF-bb pg/ml 0.751 0.290 0.458 0.894 0.356 1.357
    logHu MIP-1b pg/ml 0.709 0.444 0.508 -0.882 0.161 -0.991
    logHu RANTES pg/ml 0.748 0.252 0.866 0.166 0.823 -0.335
    loghu TNF-a pg/ml 0.787 0.127 0.143 -1.020 0.694 -0.162
    loghu VEGF pg/ml 0.400 0.558 0.389 -0.793 0.967 0.030
    sgp130 µg/ml -18.182 0.003 -24.159 0.003 0.212 -5.882
    logHu-sIL-6R pg/ml 0.118 -2.590 0.023 -5.922 0.679 0.590
    logHu-sTNFRI pg/ml 0.302 -1.284 0.843 -0.306 0.566 0.591
    logHu-sTNFRII pg/ml 0.719 -0.519 0.064 1.210 0.390 0.775
    age 0.139 0.039 0.064 -0.073 0.444 0.019
    Duretion of disease 0.228 0.041 0.221 -0059 0.414 0.033
    WBC 0.173 0.000 0.434 0.000 0.057 0.000
    DAS28-CRP 0.011 0.608 0.689 0.165 0.005 0.845
    VAS 0.328 0.013 0.810 0.005 0.419 0.011
    CRP 0.993 0.001 0.939 -0.009 0.019 0.342
    RF 0.121 0.002 0.995 0.000 0.015 0.008
    Swollen joint count 0.015 0.123 0.193 0.182 0.012 0.218
    Tender joint count 0.014 0.123 0.363 0.137 0.046 0.147
    Stage 0.237 0.328 0.352 0.651 0.615 -0.158
    Class 0.459 0.481 0.806 -0.201 0.403 0.438
  • A multivariable model was examined as a prediction biomarker for remission and non-remission by phased multiple forward logistic regression analysis based on serum concentration of cytokines/chemokines/soluble receptors in patients prior to administration of tocilizumab. Tables 12 and 13 show optimal combinations of prediction biomarkers for remission and non-remission found based on phased multiple forward logistic regression analysis and ROC curves. It was found from the results of analysis that sgp130, logIP-10, logsTNFRII and logIL-6 can be prediction biomarkers for determining with high precision whether remission is reached for naive patients who received tocilizumab therapy (p = 0.0004) (Table 11a). Further, it was found that logIL-7 (p = 0.0003), logIL-1β (p = 0.0005) or logMCP-1 (p = 0.0004), in combination with sgp130, logIP-10, and logsTNFRII, can be a prediction biomarker for determining with high precision whether remission is reached for naive patients who received anti-IL-6 therapy (tocilizumab therapy) (Tables 12b-12d).
  • Further, it was found that a combination of sgp130, log IP-10, log sTNFRII and log IL-6 can be a prediction biomarker for determining whether remission is reached for switch patients who received tocilizumab therapy (p = 0.002) (Table 13a). Furthermore, it was also found that a combination of sgp130,log IP-10,log sTNFRII and log IL-1β can also be a predication biomarker for determining with high precision whether remission is reached (p = 0.003) (Table 13b).
  • Meanwhile, p value was 0.257 for biomarker groups for predicting and determining the possibility of remission found based on the ROC curve and multiple logistic regression analysis obtained in tocilizumab therapy for naive patients who received etanercept therapy, thus demonstrating that this biomarker group cannot predict whether remission is reached (Table 14). Meanwhile, it was demonstrated that a combination of DAS-28 value prior to therapy (0wDAS-28), log VEGF, and log PDGF-bb can also predict and determine the possibility of remission to a certain extent, as shown in Table 15, by another multiple logistic regression analysis. Furthermore, it was found that a combination of log IL-9 and log TNF-α can also predict and determine the possibility of remission to a certain extent without using DAS-28 value (0wDAS-28) for naive patients who received etanercept therapy as shown in Table 16. That is, it is suggested that the pathology of rheumatoid arthritis patients is diverse, and a biomarker for predicting and determining the possibility of remission is different for patients to whom IL-6 inhibition is effective and patients for whom TNF-α inhibition is effective. [Table 12]
    Tocilizumab naïve Multiple logistic regression analysis
    multiple logistic analysis, Objective variable:remission vs non-remission
    a. Whole Model Test p=0.0004
    Parameter Estimates
    Term Estimates p value(Prob>ChiSq)
    Intercept -5,092 0,466
    sgp130 -36,648 0,001
    logHu IP-10 -4,004 0,007
    logHu-sTNFRII 5,632 0,016
    logHu IL-6 1,658 0,034
    Figure imgb0021
    Area Under
    Curve=0.85009 [Table 13]
    b. Whole Model Test p=0.0003
    Parameter Estimates
    Term Estimates p value(Prob>ChiSq)
    Intercept -3,467 0,621
    sgp130 -42,849 0,001
    logHu IP-10 -4,430 0,005
    logHu-sTNFRII 5,736 0,017
    logHu IL-7 2,705 0,035
    Figure imgb0022
    Area Under
    Curve=0.84832
    c. Whole Model Test p=0.0004
    Parameter Estimates
    Term Estimates p value(Prob>ChiSq)
    Intercept -2,834 0,684
    sgp130 -38,721 0,001
    logHu IP-10 -4,664 0,007
    logHu-sTNFRII 5,369 0,020
    logHu MCP-1 2,502 0,040
    Figure imgb0023
    [Table 13]
    d. Whole Model Test p=0.0005
    Parameter Estimates
    Term Estimates p value(Prob>ChiSq)
    Intercept -1,269 0,853
    sgp130 -39,538 0,001
    logHu IP-10 -3,807 0,008
    logHu-sTNFRII 5,086 0,026
    logHu IL-1b 1,647 0,050
    Figure imgb0024
    [Table 13]
    Tocilizumab switch Multiple logistic regression analysis multiple logistic analysis, Objective variable: remission vs non-remission
    a. Whole Model Test p=0.0020
    Parameter Estimates
    Term Estimates p value(Prob>ChiSq)
    Intercept -10,935 0,190
    sgp130# -29,051 0,020
    logHu IP-10 4,466 0,060
    logHu-sTNFRII 2,067 0,084
    logHu IL-6 -2,757 0,047
    Figure imgb0025
    Area Under Curve =0.892
    sgp130#:µg/ml [Table 14]
    b. Whole Model Test p=0.0030
    Parameter Estimates
    Term Estimate Prob>ChiSq
    Intercept -9,671 0,217
    sgp130# -27,150 0,026
    logHu IP-10 3,205 0,095
    logHu-sTNFRII 1,914 0,090
    logHu IL-1b -2,540 0,055
    Figure imgb0026
    [Table 14]
    Results of multiple logistic regression analysis on naive patients who received etanercept therapy by using biomarkers for predicting and determining the possibility of remission found based on results of multiple logistic regression analysis obtained from patients who received tocilizumab therapy
    Parameter Whole Model Test p=0.257
    Parameter Estimates
    Term Estimates p value(Prob>ChiSq)
    Intercept -6.489 0.179
    sgp130 -9.591 0.150
    logHu IL-6 -0.422 0.467
    logHu IP-10 0.893 0.435
    logHu-sTNFR II 1.789 0.235
    [Table 15]
    Etanercept naïve Multiple logistic regression analysis
    multiple logistic analysis, Objective variable :remission vs non-remission
    Whole Model Test p=0.0034
    Parameter Estimates
    Term Estimates p value(Prob>ChiSq)
    Intercept -19.058 0.025
    0w DAS28-CRP 1.390 0.007
    log VEGF -2.763 0.045
    log PDGF-bb 4.962 0.042
    Figure imgb0027
    Area Under
    Curve=0.8055
    Whole Model Test p=0.0071
    Parameter Estimates
    Term Estimates p value(Prob>ChiSq)
    Intercept 4.491 0.031
    0w DAS28-CRP 1.107 0.007
    log MIP-1a -1.808 0.082
    log PDGF-bb 3.930 0.069
    Figure imgb0028
    [Table 16]
    Etanercept naïve Multiple logistic regression analysis
    multiple logistic analysis, Objective variable:remission vs non-remission
    Whole Model Test p=0.0115
    Parameter Estimates
    Term Estimates p value(Prob>ChiSq)
    Intercept -1.004 0.337
    log IL-9 1.711 0.012
    log TNF-α -1.031 0.079

Claims (14)

  1. A method of predicting and determining a therapeutic effect of a biological formulation targeting an inflammatory cytokine on a rheumatoid arthritis patient, characterized in comprising the step of measuring a concentration of at least one type of determination marker selected from the group consisting of sgp130, IP-10, sTNFRI, sTNFRII, GM-CSF, IL-1β, IL-2, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12, IL-13, IL-15, Eotaxin, VEGF, MCP-1, TNF-α, IFN-γ, FGFbasic, PDGF-bb, sIL-6R, and MIP-1α in a serum collected from the rheumatoid arthritis patient prior to the administration of the biological formulation.
  2. The method of claim 1, wherein
    the method is a method of predicting and determining a possibility of remission with tocilizumab, and
    at least sgp130 is used as the determination marker.
  3. The method of determining of claim 2, wherein
    a patient to be administered with tocilizumab is a rheumatoid arthritis patient who has not received anti-cytokine therapy in the past, and
    the determination marker is a combination of (i) sgp130, (ii) IP-10, (iii) sTNFRII, and (iv) IL-6, IL-7, MCP-1 or IL-1β.
  4. The method of determining of claim 2, wherein
    a patient to be administered with tocilizumab is a rheumatoid arthritis patient who has received anti-cytokine therapy in the past, and
    the determination marker is a combination of (i) sgp130, (ii) IP-10, (iii) sTNFRII, and (iv) IL-6 or IL-1β.
  5. The method of determining of claim 1, wherein
    the method is a method of predicting and determining a possibility of remission with etanercept in a rheumatism patient who has not received anti-cytokine therapy in the past, and
    the determination marker is a combination of IL-9 and TNF-α, a combination of VEGF and PDGF-bb, or a combination of MIP-1α and PDGF-bb.
  6. The method of determining of claim 1, wherein
    the method is a method of predicting and determining a disease activity indicator after therapy with tocilizumab in a rheumatism patient who has not received anti-cytokine therapy in the past, and
    wherein the determination marker is a combination of sgp130, IL-8, Eotaxin, IP-10, sTNFRI, sTNFRII, and IL-6 or a combination of sgp130, IL-8, Eotaxin, IP-10, sTNFRI, sTNFRII, IL-6 andVEGF.
  7. The method of determining of claim 1, wherein
    the method is a method of predicting and determining a value of a disease activity indicator after therapy with tocilizumab in a rheumatism patient who has received anti-cytokine therapy in the past, and
    the determination marker is a combination of sgp130, IP-10, and GM-CSF.
  8. The method of determining of claim 1, wherein
    the method is a method of predicting and determining a value of a disease activity indicator after therapy with etanercept in a rheumatism patient who has not received anti-cytokine therapy in the past, and
    the determination marker is a combination of IL-9, TNF-α and VEGF or a combination of IL-6 and IL-13.
  9. The method of determining of claim 1, wherein
    the method is a method of predicting and determining a level of improvement in a symptom after therapy with tocilizumab in a rheumatism patient who has not received anti-cytokine therapy in the past, and
    the determination marker is a combination of IL-1β, IL-7, TNF-α, and sIL-6R.
  10. The method of determining of claim 1, wherein
    the method is a method of predicting and determining a level of improvement in a symptom after therapy with etanercept in a rheumatism patient who has not received anti-cytokine therapy in the past, and
    the determination marker is a combination of IL-2, IL-15, sIL-6R, and sTNFRI or a combination of IL-6 and IL-13.
  11. A method of selecting a more effective biological formulation for therapy in a rheumatism patient who has not received anti-cytokine therapy in the past from among biological formulations consisting of tocilizumab and etanercept, comprising:
    predicting and determining a possibility of remission with tocilizumab in accordance with the method of determining of claim 3;
    predicting and determining a possibility of remission with etanercept in accordance with the method of determining of claim 5; and
    comparing the possibility of remission with tocilizumab with the possibility of remission with etanercept that were predicted and determined in the aforementioned steps to select a biological formulation with a high possibility of remission.
  12. A method of selecting a more effective biological formulation for therapy in a rheumatism patient who has not received anti-cytokine therapy in the past from among biological formulations consisting of tocilizumab and etanercept, comprising:
    predicting and determining a disease activity indicator after therapy with tocilizumab in accordance with the method of determining of claim 6;
    predicting and determining a disease activity indicator after therapy with etanercept in accordance with the method of determining of claim 8; and
    comparing the disease activity indicator after therapy with tocilizumab with the disease activity indicator after therapy with etanercept that were predicted and determined in the aforementioned steps to select a biological formulation with a low disease activity indicator after therapy.
  13. A method of selecting a more effective biological formulation for therapy in a rheumatism patient who has not received anti-cytokine therapy in the past from among biological formulations consisting of tocilizumab and etanercept, comprising:
    predicting and determining a level of improvement in a symptom after therapy with tocilizumab in accordance with the method of determining of claim 9;
    predicting and determining a level of improvement in a symptom after therapy with etanercept in accordance with the method of determining of claim 10; and
    comparing the level of improvement in a symptom after therapy with tocilizumab with the level of improvement in a symptom after therapy with etanercept that were predicted in the aforementioned steps to select a biological formulation with a high level of improvement in a symptom after therapy.
  14. A diagnostic agent for predicting and determining a therapeutic effect due to a biological formulation targeting an inflammatory cytokine on a rheumatoid arthritis patient, comprising a reagent capable of detecting at least one type of marker selected from the group consisting of sgp130, IP-10, sTNFRI, sTNFRII, GM-CSF, IL-1β, IL-2, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12, IL-13, IL-15, Eotaxin, VEGF, MCP-1, TNF-α, IFN-γ, FGFbasic, PDGF-bb, sIL-6R, and MIP-1α.
EP14867880.8A 2013-12-04 2014-12-04 Method for predicting therapeutic effect of biological preparation on rheumatoid arthritis Active EP3088897B1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2013251152 2013-12-04
PCT/JP2014/082061 WO2015083765A1 (en) 2013-12-04 2014-12-04 Method for predicting therapeutic effect of biological preparation on rheumatoid arthritis

Publications (3)

Publication Number Publication Date
EP3088897A1 true EP3088897A1 (en) 2016-11-02
EP3088897A4 EP3088897A4 (en) 2017-10-11
EP3088897B1 EP3088897B1 (en) 2020-11-04

Family

ID=53273525

Family Applications (1)

Application Number Title Priority Date Filing Date
EP14867880.8A Active EP3088897B1 (en) 2013-12-04 2014-12-04 Method for predicting therapeutic effect of biological preparation on rheumatoid arthritis

Country Status (4)

Country Link
US (3) US20160377612A1 (en)
EP (1) EP3088897B1 (en)
JP (3) JPWO2015083765A1 (en)
WO (1) WO2015083765A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3309553A4 (en) * 2015-06-09 2019-02-13 Osaka University Method for predicting/evaluating therapeutic effect of biological preparation on rheumatoid arthritis

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP6631072B2 (en) * 2015-07-31 2020-01-15 富士通株式会社 Biological simulation system and biological simulation method
JP7033774B2 (en) * 2017-08-25 2022-03-11 国立大学法人 東京大学 Anti-cytokine antibody therapy
US20210382049A1 (en) 2018-05-15 2021-12-09 Navolab Diagnosztika Kft. Assessing responsiveness of rheumatoid arthritis patients to biological treatment
EP3570028A1 (en) 2018-05-15 2019-11-20 MDQuest Kft. Assessing responsiveness of rheumatoid arthritis patients to biological treatment
EP3623816A1 (en) * 2018-09-12 2020-03-18 GLX Analytix ApS Glx-derived molecule detection
WO2020175633A1 (en) * 2019-02-28 2020-09-03 吉崎 和幸 METHOD FOR PREDICTING AND DETERMINING EFFICACY OF TREATMENT OF RHEUMATOID ARTHRITIS USING ANTI-TNF–α AGENT
CN114594266B (en) * 2022-03-02 2022-12-27 安徽中医药大学第一附属医院(安徽省中医院) Application of M1 and M2 type macrophage factor as biomarker in diagnosis and treatment monitoring of rheumatoid arthritis

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
UA81743C2 (en) * 2000-08-07 2008-02-11 Центокор, Инк. HUMAN MONOCLONAL ANTIBODY WHICH SPECIFICALLY BINDS TUMOR NECROSIS FACTOR ALFA (TNFα), PHARMACEUTICAL MIXTURE CONTAINING THEREOF, AND METHOD FOR TREATING ARTHRITIS
WO2007038501A2 (en) * 2005-09-27 2007-04-05 The Feinstein Institute For Medical Research Rheumatoid arthritis markers
JP2009225713A (en) 2008-03-21 2009-10-08 Univ Of Tsukuba Method for judging effectiveness of infliximab
JP2010088432A (en) 2008-09-09 2010-04-22 Kansetsu Saisei Kenkyusho:Kk Polymorphism relating to effectiveness-side effect expression by tnf inhibitor therapy and use thereof
JP2011182780A (en) 2010-02-12 2011-09-22 Hubit Genomix Inc Polymorphism of efficacy and side effect expression of il-6 inhibitor treatment and use thereof
WO2011128096A1 (en) 2010-04-16 2011-10-20 Roche Diagnostics Gmbh Polymorphism markers for predicting response to interleukin-6 receptor-inhibiting monoclonal antibody drug treatment
US20130274133A1 (en) 2010-10-01 2013-10-17 Rigshospitalet Genetic variations in the interleukin-6 receptor gene as predictors of the response of patients to treatment with interleukin-6 receptor inhibitors

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3309553A4 (en) * 2015-06-09 2019-02-13 Osaka University Method for predicting/evaluating therapeutic effect of biological preparation on rheumatoid arthritis

Also Published As

Publication number Publication date
US20180372740A1 (en) 2018-12-27
US20210255183A1 (en) 2021-08-19
WO2015083765A1 (en) 2015-06-11
JP2021063826A (en) 2021-04-22
EP3088897B1 (en) 2020-11-04
JP2019219422A (en) 2019-12-26
JPWO2015083765A1 (en) 2017-03-16
EP3088897A4 (en) 2017-10-11
US20160377612A1 (en) 2016-12-29

Similar Documents

Publication Publication Date Title
US20210255183A1 (en) Method for predicting therapeutic effect of biological preparation on rheumatoid arthritis
Hastie et al. Analyses of asthma severity phenotypes and inflammatory proteins in subjects stratified by sputum granulocytes
Mitrovic et al. New markers for adult-onset Still's disease
Hastie et al. Complex association patterns for inflammatory mediators in induced sputum from subjects with asthma
Sucur et al. Chemokine signals are crucial for enhanced homing and differentiation of circulating osteoclast progenitor cells
Bellos et al. The role of interleukins in preeclampsia: a comprehensive review
Foell et al. Monitoring neutrophil activation in juvenile rheumatoid arthritis by S100A12 serum concentrations
Saha et al. Increased sputum and bronchial biopsy IL-13 expression in severe asthma
Shimauchi et al. Serum interleukin‐22 and vascular endothelial growth factor serve as sensitive biomarkers but not as predictors of therapeutic response to biologics in patients with psoriasis
Liu et al. Chemokine receptor expression on human eosinophils from peripheral blood and bronchoalveolar lavage fluid after segmental antigen challenge
Parys et al. Serum cytokine profiling in cats with acute idiopathic cystitis
Cossu et al. Earliest phase of systemic sclerosis typified by increased levels of inflammatory proteins in the serum
Meller et al. Chemokine responses distinguish chemical-induced allergic from irritant skin inflammation: memory T cells make the difference
Echigo et al. Both Th1 and Th2 chemokines are elevated in sera of patients with autoimmune blistering diseases
Silvagni et al. From bed to bench and back: TNF-α, IL-23/IL-17A, and JAK-dependent inflammation in the pathogenesis of psoriatic synovitis
WO2015112848A1 (en) Methods for detection of respiratory diseases
Aldinucci et al. CSF/serum matrix metallopeptidase‐9 ratio discriminates neuro Behçet from multiple sclerosis
Shono et al. Expression of CCR3 and CCR4 suggests a poor prognosis in mycosis fungoides and Sézary syndrome
US20210018514A1 (en) Method of predicting and determining therapeutic effect on rheumatoid arthritis due to biological formulation
Bai et al. Hierarchical cluster analysis of cytokine profiles reveals a cutaneous vasculitis-associated subgroup in dermatomyositis
Speck et al. Plasma and bronchoalveolar lavage samples in acute lung allograft rejection: the potential role of cytokines as diagnostic markers
Ferretti et al. Interleukin-31 and thymic stromal lymphopoietin expression in plasma and lymph node from Hodgkin lymphoma patients
Nilsson et al. Increased B-cell activating factor, interleukin-6, and interleukin-8 in induced sputum from primary Sjögren’s syndrome patients
Cabrera-Rivera et al. Increased TNF-α production in response to IL-6 in patients with systemic inflammation without infection
El-Gazzar et al. IL22 in Egyptian SLE patients, could it reflect disease activity, skin or renal involvement or is it only an expensive ESR?

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20160629

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAX Request for extension of the european patent (deleted)
RIC1 Information provided on ipc code assigned before grant

Ipc: G01N 33/68 20060101AFI20170530BHEP

A4 Supplementary search report drawn up and despatched

Effective date: 20170911

RIC1 Information provided on ipc code assigned before grant

Ipc: G01N 33/68 20060101AFI20170905BHEP

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20180720

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20200605

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: YOSHIZAKI, KAZUYUKI

RIN1 Information on inventor provided before grant (corrected)

Inventor name: IWAHASHI, MITSUHIRO

Inventor name: UNO, KAZUKO

Inventor name: YOSHIZAKI, KAZUYUKI

Inventor name: YAGI, KATSUMI

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 1331490

Country of ref document: AT

Kind code of ref document: T

Effective date: 20201115

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602014072120

Country of ref document: DE

REG Reference to a national code

Ref country code: NL

Ref legal event code: MP

Effective date: 20201104

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 1331490

Country of ref document: AT

Kind code of ref document: T

Effective date: 20201104

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20210304

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20210204

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201104

Ref country code: RS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201104

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20210205

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20210204

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201104

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201104

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201104

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20210304

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201104

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201104

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG9D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201104

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201104

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201104

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201104

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201104

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201104

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201104

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602014072120

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201104

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201104

REG Reference to a national code

Ref country code: BE

Ref legal event code: MM

Effective date: 20201231

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20210805

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201104

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201204

Ref country code: AL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201104

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201204

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201104

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201231

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201104

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201231

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20210304

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201104

Ref country code: MT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201104

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201104

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20201104

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20201231

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20231220

Year of fee payment: 10

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FR

Payment date: 20231219

Year of fee payment: 10

Ref country code: DE

Payment date: 20231214

Year of fee payment: 10